1
|
Wei Z, Yuan J, Chen Q, Pang J, Shi Q, Peng B, Wei M, Wei Y, Zhang Z, Lu X, Lin X, Liang Q. Bifunctional chemokine-nanobody fusion protein enhances neutrophil recruitment to impede Acanthamoeba immune evasion. EBioMedicine 2025; 115:105685. [PMID: 40222104 PMCID: PMC12013128 DOI: 10.1016/j.ebiom.2025.105685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Acanthamoeba keratitis (AK) is a severe infectious disease that causes serious visual impairment and low quality of life. This study aims to investigate the immune landscape in AK, with the goal of improving treatment outcomes through immunotherapy. METHODS We conducted single-cell transcriptome sequencing on corneal tissues from nine patients (3 AK patients, 3 patients with fungal keratitis and 3 patients with bacterial keratitis). Bioinformatic analysis calculated the cell subsets and their proportions within different infectious keratitis. CellChat analysis elucidated the differential expression of chemokines in keratitis. After that, screening amebic nano-antibodies, synthesizing antibody-chemokine fusion proteins, and validated their affinity and chemotactic abilities in vitro and in vivo. And assessing of the therapeutic efficacy of antibody-chemokine fusion proteins. FINDINGS The UMAP plot demonstrated the 13 major cell clusters in infectious keratitis. Compared with non-AK group, the neutrophil proportion of AK group is markedly reduced. Cell communication indicated a diminished CXCL pathway in AK. Acanthamoeba-specific antibodies were obtained by screening a natural antibody library derived from alpacas. The amoeba-specific antibodies were conjugated with the CXCL1 chemokine, and this fusion protein exhibited robust binding affinity to Acanthamoeba and chemotactic capacity both in vitro and in vivo. Furthermore, in vivo animal investigations indicated that the fusion protein presented excellent therapeutic effect and could effectively eliminate the Acanthamoeba burden. INTERPRETATION This study revealed an immune evasion mechanism employed by Acanthamoeba and offered a therapeutic approach. It presents promising potential for enhancing the treatment of infectious diseases by targeting and overcoming challenges posed by immune evasion. FUNDING This work was funded by National Natural Science Foundation of China (grant number 82171017 and 82471041) and the Beijing Municipal Public Welfare Development and Reform Pilot Project for Medical Research Institutes (PWD&RPP-MRI, JYY2023-6).
Collapse
Affiliation(s)
- Zhenyu Wei
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Jianlong Yuan
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Qiankun Chen
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Jinding Pang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Qingquan Shi
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Bo Peng
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Mingda Wei
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Yuan Wei
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Zhibao Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Xinxin Lu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China
| | - Xin Lin
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Qingfeng Liang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100005, China.
| |
Collapse
|
2
|
Xu K, Zhu S, Xu F, Yang J, Deng B, Su D, Ma J, Zu M, Lin Y, Pei T, Zhu Y, Wang L, Liu D, Duan Q, Xu J, Pan Z, Tao J, Hou Z. Toxoplasma gondii induces MST2 phosphorylation mediating the activation of hippo signaling pathway to promote apoptosis and lung tissue damage. iScience 2024; 27:111312. [PMID: 39640582 PMCID: PMC11618000 DOI: 10.1016/j.isci.2024.111312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/23/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is an intracellular parasite, and its regulation of host cell apoptosis directly affects its parasitism. Studies link T. gondii-induced apoptosis to abnormal expression of mammalian STE20-like protein kinase 2 (MST2), but its precise role remains unclear. In this study, the regulatory roles in apoptosis and pathogenicity of T. gondii infection were identified in vitro and in vivo. Simultaneously, MST2 and Hippo signaling pathway activation induced by T. gondii were evaluated. MST2 overexpression and knockout were used to assess its regulatory role in apoptosis and Hippo signaling pathway. Results showed that T. gondii induced apoptosis and lung damage, with Hippo signaling pathway activation via MST2 phosphorylation. MST2 was demonstrated to regulate apoptosis and Hippo signaling pathway. Notably, MST2 knockout hindered the T. gondii-induced apoptosis and weakened Hippo signaling pathway activation. MST2 is an important target for T. gondii to control host cell fate and modulate immune responses.
Collapse
Affiliation(s)
- Kangzhi Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Shifan Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Fan Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Jin Yang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225000, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou 225000, China
| | - Dingzeyang Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Jing Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Mingyue Zu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Yifan Lin
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Tianxu Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Yuyang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Lele Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Qiangde Duan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| |
Collapse
|
3
|
Barik S, Andrews J. Host-Parasite Interactions in Toxoplasma gondii-Infected Cells: Roles of Mitochondria, Microtubules, and the Parasitophorous Vacuole. Int J Mol Sci 2024; 25:13459. [PMID: 39769222 PMCID: PMC11677533 DOI: 10.3390/ijms252413459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
An intracellular protozoan, the Apicomplexan parasite Toxoplasma gondii (T. gondii) infects nucleated cells, in which it triggers the formation of a specialized membrane-confined cytoplasmic vacuole, named the parasitophorous vacuole (PV). One of the most prominent events in the parasite's intracellular life is the congregation of the host cell mitochondria around the PV. However, the significance of this event has remained largely unsolved since the parasite itself possesses a functional mitochondrion, which is essential for its replication. Here, we explore several fundamental aspects of the interaction between the PV and the host cell mitochondria. They include the detailed features of the congregation, the nature and mechanism of the mitochondrial travel to the PV, and the potential significance of the migration and congregation. Using a combination of biochemical assays, high-resolution imaging, and RNAi-mediated knockdown, we show that: (i) mitochondrial travel to the PV starts very early in parasite infection, as soon as the smallest PV takes shape; (ii) the travel utilizes the contractile microtubular network of the host cell; and (iii) near the end of the parasitic life cycle, when most PVs have reached their largest sustainable size and are about to lyse in order to release the progeny parasites, the associated mitochondria change their usual elongated shape to small spheres, apparently resulting from increased fission. Intriguingly, despite the well-known mitochondrial role as a major producer of cellular ATP, the parasite does not seem to use cellular mitochondrial ATP. Together, these findings may serve as foundations for future research in host-parasite interaction, particularly in the elucidation of its mechanisms, and the possible development of novel antiparasitic drug regimens.
Collapse
|
4
|
Asgari Q, Rajabi F, Sajadian F, Bahreini MS, Arefkhah N. Toxoplasma gondii infection in patients with brain tumors in Southern Iran: a case-control study. J Parasit Dis 2023; 47:291-296. [PMID: 37193506 PMCID: PMC10182190 DOI: 10.1007/s12639-022-01541-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/12/2022] [Indexed: 02/06/2023] Open
Abstract
Considerable evidence points to a dominant role of inflammation in tumor pathology. The biological response of the immune system can be triggered by Toxoplasma gondii as a common brain-tropic parasite. The aim of this study was to investigate an association between Toxoplasma infection and brain tumors. This case-control study was performed on sera of brain tumor patients (n = 124) and age- and sex-matched control subjects (n = 124) in Southern Iran. Data related to tumor site and type were collected during sample collection. Anti-Toxoplasma IgG was assessed by enzyme-linked immunosorbent assay (ELISA). Seroprevalence anti-Toxoplasma IgG was significantly higher in brain tumor patients 30.6% (38/124) compared with 12.1% (15/124) of the healthy controls (OR 3.211; 95% CI 1.658 to 6.219; p = 0.001). The highest seroprevalence was detected in patients with ependymoma (100%), followed by glioblastoma (83%), pituitary adenoma (47.3%), astrocytoma (27.2%), schwannoma (23%), and meningioma (22.6%). The parasite infection was correlated to brain tumor's location i.e., the patients with frontal lobe and sella region tumors had higher seropositivity compared with others (P < 0.05). The higher prevalence of Toxoplasma infection among patients with brain tumor compared with the control group indicates a probable association between the infection and brain tumors.
Collapse
Affiliation(s)
- Qasem Asgari
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farshad Rajabi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fataneh Sajadian
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohammad Saleh Bahreini
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasir Arefkhah
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
5
|
Functional Characterization of 15 Novel Dense Granule Proteins in Toxoplasma gondii Using the CRISPR-Cas9 System. Microbiol Spectr 2023; 11:e0307822. [PMID: 36515555 PMCID: PMC9927372 DOI: 10.1128/spectrum.03078-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The analysis of the subcellular localization and function of dense granule proteins (GRAs) is of central importance for the understanding of host-parasite interaction and pathogenesis of Toxoplasma gondii infection. Here, we identified 15 novel GRAs and used C-terminal endogenous gene tagging to determine their localization at the intravacuolar network (IVN), parasitophorous vacuole (PV), or PV membrane (PVM) in the tachyzoites and at the periphery of the bradyzoites-containing cysts. The functions of the 15 gra genes were examined in type I RH strain and 5 of these gra genes were also evaluated in the cyst-forming type II Pru strain. The 15 novel gra genes were successfully disrupted by using CRISPR-Cas9 mediated homologous recombination and the results showed that 13 gra genes were not individually essential for T. gondii replication in vitro or virulence in mice during acute and chronic infection. Intriguingly, deletion of TGME49_266410 and TGME49_315910 in both RH and Pru strains decreased the parasite replication in vitro and attenuated its virulence, and also reduced the cyst-forming ability of the Pru strain in mice during chronic infection. Comparison of the transcriptomic profiles of the 15 gra genes suggests that they may play roles in other life cycle stages and genotypes of T. gondii. Taken together, our findings improve the understanding of T. gondii pathogenesis and demonstrate the involvement of two novel GRAs, TGME49_266410 and TGME49_315910, in the parasite replication and virulence. IMPORTANCE Dense granule proteins (GRAs) play important roles in Toxoplasma gondii pathogenicity. However, the functions of many putative GRAs have not been elucidated. Here, we found that 15 novel GRAs are secreted into intravacuolar network (IVN), parasitophorous vacuole (PV), or PV membrane (PVM) in tachyzoites and are located at the periphery of the bradyzoite-containing cysts. TGME49_266410 and TGME49_315910 were crucial to the growth of RH and Pru strains in vitro. Deletion of TGME49_266410 and TGME49_315910 attenuated the parasite virulence in mice. However, disruption of other 13 gra genes did not have a significant impact on the proliferation and pathogenicity of T. gondii in vitro or in vivo. The marked effects of the two novel GRAs (TGME49_266410 and TGME49_315910) on the in vitro growth and virulence of T. gondii are notable and warrant further elucidation of the temporal and spatial dynamics of translocation of these two novel GRAs and how do they interfere with host cell functions.
Collapse
|
6
|
Overview of Apoptosis, Autophagy, and Inflammatory Processes in Toxoplasma gondii Infected Cells. Pathogens 2023; 12:pathogens12020253. [PMID: 36839525 PMCID: PMC9966443 DOI: 10.3390/pathogens12020253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular parasite. During the parasitic invasion, T. gondii creates a parasitophorous vacuole, which enables the modulation of cell functions, allowing its replication and host infection. It has effective strategies to escape the immune response and reach privileged immune sites and remain inactive in a controlled environment in tissue cysts. This current review presents the factors that affect host cells and the parasite, as well as changes in the immune system during host cell infection. The secretory organelles of T. gondii (dense granules, micronemes, and rhoptries) are responsible for these processes. They are involved with proteins secreted by micronemes and rhoptries (MIC, AMA, and RONs) that mediate the recognition and entry into host cells. Effector proteins (ROP and GRA) that modify the STAT signal or GTPases in immune cells determine their toxicity. Interference byhost autonomous cells during parasitic infection, gene expression, and production of microbicidal molecules such as reactive oxygen species (ROS) and nitric oxide (NO), result in the regulation of cell death. The high level of complexity in host cell mechanisms prevents cell death in its various pathways. Many of these abilities play an important role in escaping host immune responses, particularly by manipulating the expression of genes involved in apoptosis, necrosis, autophagy, and inflammation. Here we present recent works that define the mechanisms by which T. gondii interacts with these processes in infected host cells.
Collapse
|
7
|
Effects of different drugs and hormone treatment on Toxoplasma gondii glutathione S-transferase 2. Parasit Vectors 2022; 15:461. [PMID: 36510329 PMCID: PMC9746104 DOI: 10.1186/s13071-022-05589-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Glutathione S-transferase (GST) in eukaryotic organisms has multiple functions such as detoxifying endogenous/exogenous harmful substances to protect cells from oxidative damage, participating in sterol synthesis and metabolism, and regulating signaling pathways. Our previous work identified an important GST protein in Toxoplasma that contributes to vesicle trafficking called TgGST2, the deletion of which significantly reduces the virulence of the parasite. Meanwhile, we considered that TgGST2 may also play a role in other pathways of parasite life activities. METHODS The tertiary structures of TgGST2 as well as estradiol (E2) and progesterone (P4) were predicted by trRosetta and Autodock Vina software, the binding sites were analyzed by PyMol's GetBox Plugin, and the binding capacity was evaluated using Discovery Studio plots software. We examined the influence of E2 and P4 on TgGST2 via glutathione S-transferase enzyme activity and indirect immunofluorescence assay (IFA) and through the localization observation of TgGST2 to evaluate its response ability in different drugs. RESULTS TgGST2 could bind to exogenous E2 and P4, and that enzymatic activity was inhibited by the hormones in a concentration-dependent manner. Upon P4 treatment, the localization of TgGST2 changed from Golgi and vesicles to hollow circles, leading to abnormal localization of the molecular transporter Sortilin (VPS10) and microneme proteins (M2AP and MIC2), which ultimately affect the parasite life activities, but E2 had no significant effect. Moreover, diverse types of drugs had divergent effects on TgGST2, among which treatment with antifungal agents (voriconazole and clarithromycin), anticarcinogens (KU-60019, WYE-132 and SH5-07) and coccidiostats (dinitolmide and diclazuril) made the localization of TgGST2 appear in different forms, including dots, circles and rod shaped. CONCLUSIONS Our study shows that TgGST2 plays a role in sterol treatment and can be affected by P4, which leads to deficient parasite motility. TgGST2 exerts divergent effects in response to the different properties of the drugs themselves. Its responsiveness to diverse drugs implies a viable target for the development of drugs directed against Toxoplasma and related pathogenic parasites.
Collapse
|
8
|
Węglińska L, Bekier A, Trotsko N, Kaproń B, Plech T, Dzitko K, Paneth A. Inhibition of Toxoplasma gondii by 1,2,4-triazole-based compounds: marked improvement in selectivity relative to the standard therapy pyrimethamine and sulfadiazine. J Enzyme Inhib Med Chem 2022; 37:2621-2634. [PMID: 36165032 PMCID: PMC9518248 DOI: 10.1080/14756366.2022.2112576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A safer treatment for toxoplasmosis would be achieved by improving the selectivity profile of novel chemotherapeutics compared to the standard therapy pyrimethamine (PYR) and sulfadiazine (SDZ). We previously reported on the identification of the compounds with imidazole-thiosemicarbazide scaffold as potent and selective anti-Toxoplasma gondii (T. gondii) agents. In our current research, we report on the optimisation of this chemical scaffold leading to the discovery cyclic analogue 20 b with s-triazole core structure. This compound displayed prominent CC30 to IC50 selectivity index (SI) of 70.72, making it 160-fold more selective than SDZ, 11-fold more selective than PYR, and 4-fold more selective than trimethoprim (TRI). Additionally, this compound possesses prerequisite drug-like anti-Toxoplasma properties to advance into preclinical development; it showed ability to cross the BBB, did not induce genotoxic and haemolytic changes in human cells, and as well as it was characterised by low cellular toxicity.
Collapse
Affiliation(s)
- Lidia Węglińska
- Department of Organic Chemistry, Medical University of Lublin, Lublin, Poland
| | - Adrian Bekier
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Nazar Trotsko
- Department of Organic Chemistry, Medical University of Lublin, Lublin, Poland
| | - Barbara Kaproń
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Tomasz Plech
- Department of Pharmacology, Medical University of Lublin, Lublin, Poland
| | - Katarzyna Dzitko
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Agata Paneth
- Department of Organic Chemistry, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
9
|
Rosini AM, Teixeira SC, Milian ICB, Silva RJ, de Souza G, Luz LC, Gomes AO, Mineo JR, Mineo TWP, Ferro EAV, Barbosa BF. LPS-mediated activation of TLR4 controls Toxoplasma gondii growth in human trophoblast cell (BeWo) and human villous explants in a dependent-manner of TRIF, MyD88, NF-κB and cytokines. Tissue Cell 2022; 78:101907. [DOI: 10.1016/j.tice.2022.101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/22/2022] [Accepted: 08/22/2022] [Indexed: 02/07/2023]
|
10
|
Ulahannan N, Cutler R, Doña-Termine R, Simões-Pires CA, Wijetunga NA, Croken MM, Johnston AD, Kong Y, Maqbool SB, Suzuki M, Greally JM. Genomic insights into host and parasite interactions during intracellular infection by Toxoplasma gondii. PLoS One 2022; 17:e0275226. [PMID: 36178892 PMCID: PMC9524707 DOI: 10.1371/journal.pone.0275226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
To gain insights into the molecular interactions of an intracellular pathogen and its host cell, we studied the gene expression and chromatin states of human fibroblasts infected with the Apicomplexan parasite Toxoplasma gondii. We show a striking activation of host cell genes that regulate a number of cellular processes, some of which are protective of the host cell, others likely to be advantageous to the pathogen. The simultaneous capture of host and parasite genomic information allowed us to gain insights into the regulation of the T. gondii genome. We show how chromatin accessibility and transcriptional profiling together permit novel annotation of the parasite's genome, including more accurate mapping of known genes and the identification of new genes and cis-regulatory elements. Motif analysis reveals not only the known T. gondii AP2 transcription factor-binding site but also a previously-undiscovered candidate TATA box-containing motif at one-quarter of promoters. By inferring the transcription factor and upstream cell signaling responses involved in the host cell, we can use genomic information to gain insights into T. gondii's perturbation of host cell physiology. Our resulting model builds on previously-described human host cell signalling responses to T. gondii infection, linked to induction of specific transcription factors, some of which appear to be solely protective of the host cell, others of which appear to be co-opted by the pathogen to enhance its own survival.
Collapse
Affiliation(s)
- Netha Ulahannan
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Ronald Cutler
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Reanna Doña-Termine
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Claudia A. Simões-Pires
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - N. Ari Wijetunga
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Matthew McKnight Croken
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Andrew D. Johnston
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Yu Kong
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Shahina B. Maqbool
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Masako Suzuki
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - John M. Greally
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| |
Collapse
|
11
|
Guo G, Cui J, Song L, Tang L, Fan S, Shen B, Fang R, Hu M, Zhao J, Zhou Y. Activation of NF-κB signaling by the dense granule protein GRA15 of a newly isolated type 1 Toxoplasma gondii strain. Parasit Vectors 2022; 15:347. [PMID: 36175964 PMCID: PMC9523984 DOI: 10.1186/s13071-022-05429-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Background It has been reported that the NF-κB pathway, an important component of host defense system against pathogens infections, can be differentially modulated by different Toxoplasma gondii strains, depending on the polymorphism of the GRA15 protein. The recently isolated Toxoplasma strain T.gHB1 is a type 1 (ToxoDB#10) strain but shows different virulence determination mechanisms compared to the classic type 1 strains like RH (ToxoDB#10). Therefore, it is worth investigating whether the T.gHB1 strain (ToxoDB#10) affects the host NF-κB signaling pathway. Methods The effects of T.gHB1 (ToxoDB#10) on host NF-κB pathway were investigated in HEK293T cells. The GRA15 gene product was analyzed by bioinformatics, and its effect on NF-κB activation was examined by Western blotting and nuclear translocation of p65. Different truncations of T.gHB1 GRA15 were constructed to map the critical domains for NF-κB activation. Results We demonstrated that the NF-κB pathway signaling pathway could be activated by the newly identified type 1 T.gHB1 strain (ToxoDB#10) of Toxoplasma, while the classic type 1 strain RH (ToxoDB#10) did not. T.gHB1 GRA15 possesses only one transmembrane region with an extended C terminal region, which is distinct from that of classic type 1 (ToxoDB#10) and type 2 (ToxoDB#1) strains. T.gHB1 GRA15 could clearly induce IκBα phosphorylation and p65 nuclear translocation. Dual luciferase assays in HEK293T cells revealed a requirement for 194–518 aa of T.gHB1 GRA15 to effectively activate NF-κB. Conclusions The overall results indicated that the newly isolated type 1 isolate T.gHB1 (ToxoDB#10) had a unique GRA15, which could activate the host NF-κB signaling through inducing IκBα phosphorylation and p65 nuclear translocation. These results provide new insights for our understanding of the interaction between Toxoplasma parasites and its hosts. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Guanghao Guo
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Jianmin Cui
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Lindong Song
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Lvqing Tang
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Sijie Fan
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Bang Shen
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Min Hu
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Junlong Zhao
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Yanqin Zhou
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China. .,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
12
|
Obed C, Wu M, Chen Y, An R, Cai H, Luo Q, Yu L, Wang J, Liu F, Shen J, Du J. Toxoplasma gondii dense granule protein 3 promotes endoplasmic reticulum stress-induced apoptosis by activating the PERK pathway. Parasit Vectors 2022; 15:276. [PMID: 35918751 PMCID: PMC9344675 DOI: 10.1186/s13071-022-05394-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background Toxoplasma gondii is a neurotropic single-celled parasite that can infect mammals, including humans. Central nervous system infection with T. gondii infection can lead to Toxoplasma encephalitis. Toxoplasma infection can cause endoplasmic reticulum (ER) stress and unfolded protein response (UPR) activation, which ultimately can lead to apoptosis of host cells. The dense granule protein GRA3 has been identified as one of the secretory proteins that contribute to the virulence of T. gondii; however, the mechanism remains enigmatic. Methods The expression of the GRA3 gene in RH, ME49, Wh3, and Wh6 strains was determined using quantitative real-time polymerase chain reaction (qRT–PCR). pEGFP-GRA3Wh6 was constructed by inserting Chinese 1 Wh6 GRA3 (GRA3Wh6) cDNA into a plasmid encoding the enhanced GFP. Mouse neuro2a (N2a) cells were transfected with either pEGFP or pEGFP-GRA3Wh6 (GRA3Wh6) and incubated for 24–36 h. N2a cell apoptosis and ER stress-associated proteins were determined using flow cytometry and immunoblotting. Furthermore, N2a cells were pretreated with GSK2656157 (a PERK inhibitor) and Z-ATAD-FMK (a caspase-12 inhibitor) before GRA3Wh6 transfection, and the effect of the inhibitors on GRA3Wh6-induced ER stress and apoptosis were investigated. Results GRA3 gene expression was higher in the less virulent strains of type II ME49 and type Chinese 1 Wh6 strains compared with the virulent strains of type I RH strain and type Chinese 1 Wh3 strain. Transfection with GRA3Wh6 plasmid induced neuronal apoptosis and increased the expression of GRP78, p-PERK, cleaved caspase-12, cleaved caspase-3, and CHOP compared with the control vector. Pretreatment with GSK2656157 and Z-ATAD-FMK decreased apoptosis in N2a cells, and similarly, ER stress- and apoptosis-associated protein levels were significantly decreased. Conclusion GRA3 induces neural cell apoptosis via the ER stress signaling pathway, which could play a role in toxoplasmic encephalitis. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Cudjoe Obed
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China.,Department of Microbiology & Immunology School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Minmin Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Ying Chen
- The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,School of Nursing, Anhui Medical University, Hefei, 230032, China
| | - Ran An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Haijian Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Qingli Luo
- The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Li Yu
- The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Jie Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Fang Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Jilong Shen
- The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China. .,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Jian Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, Anhui Medical University, Hefei, 230032, China. .,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
13
|
Kinetic Tracking of Plasmodium falciparum Antigens on Infected Erythrocytes with a Novel Reporter of Protein Insertion and Surface Exposure. mBio 2022; 13:e0040422. [PMID: 35420481 PMCID: PMC9239273 DOI: 10.1128/mbio.00404-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Intracellular malaria parasites export many proteins into their host cell, inserting several into the erythrocyte plasma membrane to enable interactions with their external environment. While static techniques have identified some surface-exposed proteins, other candidates have eluded definitive localization and membrane topology determination. Moreover, both export kinetics and the mechanisms of membrane insertion remain largely unexplored. We introduce Reporter of Insertion and Surface Exposure (RISE), a method for continuous nondestructive tracking of antigen exposure on infected cells. RISE utilizes a small 11-amino acid (aa) HiBit fragment of NanoLuc inserted into a target protein and detects surface exposure through high-affinity complementation to produce luminescence. We tracked the export and surface exposure of CLAG3, a parasite protein linked to nutrient uptake, throughout the Plasmodiumfalciparum cycle in human erythrocytes. Our approach revealed key determinants of trafficking and surface exposure. Removal of a C-terminal transmembrane domain aborted export. Unexpectedly, certain increases in the exposed reporter size improved the luminescence signal, but other changes abolished the surface signal, revealing that both size and charge of the extracellular epitope influence membrane insertion. Marked cell-to-cell variation with larger inserts containing multiple HiBit epitopes suggests complex regulation of CLAG3 insertion at the host membrane. Quantitative, continuous tracking of CLAG3 surface exposure thus reveals multiple factors that determine this protein’s trafficking and insertion at the host erythrocyte membrane. The RISE assay will enable study of surface antigens from divergent intracellular pathogens.
Collapse
|
14
|
Toxoplasma gondii infection/exposure and the risk of brain tumors: A systematic review and meta-analysis. Cancer Epidemiol 2022; 77:102119. [DOI: 10.1016/j.canep.2022.102119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 01/10/2023]
|
15
|
Doherty CM, Romero AD, Denkers EY. Impact of IFN-y and CD40 signalling on Toxoplasma gondii cyst formation in differentiated Neuro-2a neuroblastoma cells. Parasite Immunol 2022; 44:e12897. [PMID: 34762755 PMCID: PMC10809350 DOI: 10.1111/pim.12897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/02/2021] [Accepted: 11/07/2021] [Indexed: 02/01/2023]
Abstract
Signalling by IFN-y and CD40 is known to trigger anti-microbial activity in macrophages infected with Toxoplasma gondii, but their effects on infected neurons are less well known. Here, we compared how stimulation with IFN-y and an agonistic anti-CD40 mAb impacts infection and cyst formation in the mouse neuroblastoma cell line Neuro-2a relative to bone marrow-derived macrophages. Both IFN-y and CD40 mAb decreased cyst emergence in Neuro-2a cells. In macrophages, these stimuli decreased infection, but had no impact on infection in the neuroblastoma cell line. Resistance to killing in Neuro-2a cells may explain why neurons preferentially harbour parasites during chronic infection in the brain.
Collapse
Affiliation(s)
- Claire M Doherty
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Alicia D Romero
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Eric Y Denkers
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
16
|
Daher D, Shaghlil A, Sobh E, Hamie M, Hassan ME, Moumneh MB, Itani S, El Hajj R, Tawk L, El Sabban M, El Hajj H. Comprehensive Overview of Toxoplasma gondii-Induced and Associated Diseases. Pathogens 2021; 10:pathogens10111351. [PMID: 34832507 PMCID: PMC8625914 DOI: 10.3390/pathogens10111351] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 12/03/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a prevalent protozoan parasite of medical and veterinary significance. It is the etiologic agent of toxoplasmosis, a neglected disease in which incidence and symptoms differ between patients and regions. In immunocompetent patients, toxoplasmosis manifests as acute and chronic forms. Acute toxoplasmosis presents as mild or asymptomatic disease that evolves, under the host immune response, into a persistent chronic disease in healthy individuals. Chronic toxoplasmosis establishes as latent tissue cysts in the brain and skeletal muscles. In immunocompromised patients, chronic toxoplasmosis may reactivate, leading to a potentially life-threatening condition. Recently, the association between toxoplasmosis and various diseases has been shown. These span primary neuropathies, behavioral and psychiatric disorders, and different types of cancer. Currently, a direct pre-clinical or clinical molecular connotation between toxoplasmosis and most of its associated diseases remains poorly understood. In this review, we provide a comprehensive overview on Toxoplasma-induced and associated diseases with a focus on available knowledge of the molecular players dictating these associations. We will also abridge the existing therapeutic options of toxoplasmosis and highlight the current gaps to explore the implications of toxoplasmosis on its associated diseases to advance treatment modalities.
Collapse
Affiliation(s)
- Darine Daher
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Ahmad Shaghlil
- Department of Biology, Faculty of Sciences, R. Hariri Campus, Lebanese University, Beirut 1107 2020, Lebanon; (A.S.); (E.S.)
| | - Eyad Sobh
- Department of Biology, Faculty of Sciences, R. Hariri Campus, Lebanese University, Beirut 1107 2020, Lebanon; (A.S.); (E.S.)
| | - Maguy Hamie
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Malika Elhage Hassan
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Mohamad Bahij Moumneh
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Shaymaa Itani
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, Beirut 1107 2809, Lebanon;
| | - Lina Tawk
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon;
| | - Marwan El Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
- Correspondence: ; Tel.: +961–1-350000 (ext. 4897)
| |
Collapse
|
17
|
Menati Rashno M, Mehraban H, Naji B, Radmehr M. Microbiome in human cancers. Access Microbiol 2021; 3:000247. [PMID: 34888478 PMCID: PMC8650843 DOI: 10.1099/acmi.0.000247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
A microbiome is defined as the aggregate of all microbiota that reside in human digestive system and other tissues. This microbiota includes viruses, bacteria, fungi that live in various human organs and tissues like stomach, guts, oesophagus, mouth cavity, urinary tract, vagina, lungs, and skin. Almost 20 % of malignant cancers worldwide are related to microbial infections including bacteria, parasites, and viruses. The human body is constantly being attacked by microbes during its lifetime and microbial pathogens that have tumorigenic effects in 15-20 % of reported cancer cases. Recent scientific advances and the discovery of the effect of microbes on cancer as a pathogen or as a drug have significantly contributed to our understanding of the complex relationship between microbiome and cancer. The aim of this study is to overview some microbiomes that reside in the human body and their roles in cancer.
Collapse
Affiliation(s)
| | - Hamed Mehraban
- Department of Biology, Payame Noor University (PNU), Tehran, Iran
| | - Behnaz Naji
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mohadeseh Radmehr
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
18
|
Staurengo-Ferrari L, Sanfelice RADS, de Souza JB, Assolini JP, Dos Santos DP, Cataneo AHD, Bordignon J, Conchon-Costa I, da Costa IN, Fernandes GSA. Impact of Toxoplasma gondii infection on TM3 Leydig cells: Alterations in testosterone and cytokines levels. Acta Trop 2021; 220:105938. [PMID: 33932363 DOI: 10.1016/j.actatropica.2021.105938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Leydig cells play pivotal roles in eliciting male characteristics by producing testosterone and any damage to these cells can compromise male fertility Toxoplasma gondii (T. gondii) is an intracellular parasite capable to invade any nucleated cell, including cells from male reproductive system. Herein, we evaluated the capacity of RH strain of T. gondii to infect TM3 Leydig cells and the impact of this infection on testosterone and inflammatory mediators production. We first, by performing adherence, infection, and intracellular proliferation assays, we found a significant increase in the number of infected Leydig cells, peaking 48 h after the infection with T. gondii. Supernatants of TM3 infected cells exhibited, in a time-dependent manner, increased levels of testosterone as well as monocyte chemoattractant protein-1 (MCP-1) and interferon-γ (IFN-γ), which is correlated with the robust T. gondii infection. In conclusion, our study provides new insights regarding the harmful effects of T. gondii infection on male reproductive system.
Collapse
Affiliation(s)
- Larissa Staurengo-Ferrari
- Department of Pathological Sciences, State University of Londrina, Rodovia Celso Garcia Cid Pr 445 Km 380, Londrina 86057-970, Brazil
| | - Raquel Arruda da Silva Sanfelice
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer-LIDNC, Department, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Julia Bagatim de Souza
- Department of General Biology, State University of Londrina, Rodovia Celso Garcia Cid Pr 445 Km 380, Londrina 86057-970, Brazil
| | - Joao Paulo Assolini
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer-LIDNC, Department, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Dayane Priscila Dos Santos
- Department of Pathological Sciences, State University of Londrina, Rodovia Celso Garcia Cid Pr 445 Km 380, Londrina 86057-970, Brazil; Department of General Biology, State University of Londrina, Rodovia Celso Garcia Cid Pr 445 Km 380, Londrina 86057-970, Brazil
| | | | - Juliano Bordignon
- Laboratory of Molecular Virology, Instituto Carlos Chagas/Fiocruz-PR, Curitiba, Paraná Brazil
| | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer-LIDNC, Department, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Idessania Nazareth da Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer-LIDNC, Department, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | | |
Collapse
|
19
|
de Assis DRR, Pimentel PMDO, Dos Reis PVM, Rabelo RAN, Vitor RWA, Cordeiro MDN, Felicori LF, Olórtegui CDC, Resende JM, Teixeira MM, Borges MH, de Lima ME, Pimenta AMDC, Machado FS. Tityus serrulatus (Scorpion): From the Crude Venom to the Construction of Synthetic Peptides and Their Possible Therapeutic Application Against Toxoplasma gondii Infection. Front Cell Infect Microbiol 2021; 11:706618. [PMID: 34354963 PMCID: PMC8329421 DOI: 10.3389/fcimb.2021.706618] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022] Open
Abstract
Toxoplasmosis, caused by Toxoplasma gondii, is a major public concern owing to its neurotropic nature and high morbidity and mortality rates in immunocompromised patients and newborns. Current treatment for this disease is inefficient and produces side effects. Inflammatory mediators produced during T. gondii infection (e.g., cytokines and nitric oxide) are crucial in controlling parasite replication. In this context, Tityus serrulatus venom (TsV) induces the production of inflammatory mediators by immune cells. Thus, this study aimed to isolate and identify the components of TsV with potential anti-T. gondii activity. TsV was extracted from scorpions and lyophilized or loaded onto a column to obtain its fractions. TsV subfractions were obtained using chromatography, and its amino acid sequence was identified and applied to peptide design using bioinformatics tools. The C57BL/6 mice and their harvested macrophages were used to test the anti-Toxoplasma activity of TsV components and peptides. TsV and its fraction F6 attenuated the replication of tachyzoites in macrophages and induced nitric oxide and cytokine (IL-12, TNF, and IL-6) production by infected cells, without host cell toxicity. Moreover, Su6-B toxin, a subfraction of F6, demonstrated anti-T. gondii activity. The partially elucidated and characterized amino acid sequence of Sub6-B demonstrated 93% similarity with T. serrulatus 2 toxin (Ts2). Ts2 mimetic peptides (“Pep1,” “Pep2a,” and “Pep2b”) were designed and synthesized. Pep1 and Pep2a, but not Pep2b, reduced the replication of tachyzoites in macrophages. In vivo, treatment of T. gondii-infected mice with Pep1, Pep2a, or Pep2b decreased the number of cerebral cysts and did not induce hepatotoxicity in the animals. Taken together, our data show promising immunomodulatory and antiparasitic activity of TsV that could be explored and applied in future therapies for treating infectious parasitic diseases such as toxoplasmosis.
Collapse
Affiliation(s)
- Diego Rodney Rodrigues de Assis
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Pablo Victor Mendes Dos Reis
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rayane Aparecida Nonato Rabelo
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Wagner Almeida Vitor
- Department of Parasitology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Liza Figueiredo Felicori
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carlos Delfin Chávez Olórtegui
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jarbas Magalhães Resende
- Department of Chemistry, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Maria Elena de Lima
- Faculdade Santa Casa de Belo Horizonte: Programa de Pós Graduação em Medicina-Biomedicina, Belo Horizonte, Brazil
| | - Adriano Monteiro de Castro Pimenta
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Program in Health Sciences: Infectious Diseases and Tropical Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
20
|
Kellermann M, Scharte F, Hensel M. Manipulation of Host Cell Organelles by Intracellular Pathogens. Int J Mol Sci 2021; 22:ijms22126484. [PMID: 34204285 PMCID: PMC8235465 DOI: 10.3390/ijms22126484] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Pathogenic intracellular bacteria, parasites and viruses have evolved sophisticated mechanisms to manipulate mammalian host cells to serve as niches for persistence and proliferation. The intracellular lifestyles of pathogens involve the manipulation of membrane-bound organellar compartments of host cells. In this review, we described how normal structural organization and cellular functions of endosomes, endoplasmic reticulum, Golgi apparatus, mitochondria, or lipid droplets are targeted by microbial virulence mechanisms. We focus on the specific interactions of Salmonella, Legionella pneumophila, Rickettsia rickettsii, Chlamydia spp. and Mycobacterium tuberculosis representing intracellular bacterial pathogens, and of Plasmodium spp. and Toxoplasma gondii representing intracellular parasites. The replication strategies of various viruses, i.e., Influenza A virus, Poliovirus, Brome mosaic virus, Epstein-Barr Virus, Hepatitis C virus, severe acute respiratory syndrome virus (SARS), Dengue virus, Zika virus, and others are presented with focus on the specific manipulation of the organelle compartments. We compare the specific features of intracellular lifestyle and replication cycles, and highlight the communalities in mechanisms of manipulation deployed.
Collapse
Affiliation(s)
- Malte Kellermann
- Abt. Mikrobiologie, Fachbereich Biologie/Chemie, Barbarastr 11, Universität Osnabrück, 49076 Osnabrück, Germany; (M.K.); (F.S.)
| | - Felix Scharte
- Abt. Mikrobiologie, Fachbereich Biologie/Chemie, Barbarastr 11, Universität Osnabrück, 49076 Osnabrück, Germany; (M.K.); (F.S.)
| | - Michael Hensel
- Abt. Mikrobiologie, Fachbereich Biologie/Chemie, Barbarastr 11, Universität Osnabrück, 49076 Osnabrück, Germany; (M.K.); (F.S.)
- CellNanOs–Center of Cellular Nanoanalytics Osnabrück, Universität Osnabrück, Barbarastr 11, 49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-(0)-541-969-3940
| |
Collapse
|
21
|
French T, Israel N, Düsedau HP, Tersteegen A, Steffen J, Cammann C, Topfstedt E, Dieterich D, Schüler T, Seifert U, Dunay IR. The Immunoproteasome Subunits LMP2, LMP7 and MECL-1 Are Crucial Along the Induction of Cerebral Toxoplasmosis. Front Immunol 2021; 12:619465. [PMID: 33968021 PMCID: PMC8099150 DOI: 10.3389/fimmu.2021.619465] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/16/2021] [Indexed: 11/28/2022] Open
Abstract
Cell survival and function critically relies on the fine-tuned balance of protein synthesis and degradation. In the steady state, the standard proteasome is sufficient to maintain this proteostasis. However, upon inflammation, the sharp increase in protein production requires additional mechanisms to limit protein-associated cellular stress. Under inflammatory conditions and the release of interferons, the immunoproteasome (IP) is induced to support protein processing and recycling. In antigen-presenting cells constitutively expressing IPs, inflammation-related mechanisms contribute to the formation of MHC class I/II-peptide complexes, which are required for the induction of T cell responses. The control of Toxoplasma gondii infection relies on Interferon-γ (IFNγ)-related T cell responses. Whether and how the IP affects the course of anti-parasitic T cell responses along the infection as well as inflammation of the central nervous system is still unknown. To answer this question we used triple knockout (TKO) mice lacking the 3 catalytic subunits of the immunoproteasome (β1i/LMP2, β2i/MECL-1 and β5i/LMP7). Here we show that the numbers of dendritic cells, monocytes and CD8+ T cells were reduced in Toxoplasma gondii-infected TKO mice. Furthermore, impaired IFNγ, TNF and iNOS production was accompanied by dysregulated chemokine expression and altered immune cell recruitment to the brain. T cell differentiation was altered, apoptosis rates of microglia and monocytes were elevated and STAT3 downstream signaling was diminished. Consequently, anti-parasitic immune responses were impaired in TKO mice leading to elevated T. gondii burden and prolonged neuroinflammation. In summary we provide evidence for a critical role of the IP subunits β1i/LMP2, β2i/MECL-1 and β5i/LMP7 for the control of cerebral Toxoplasma gondii infection and subsequent neuroinflammation.
Collapse
Affiliation(s)
- Timothy French
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Nicole Israel
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Anne Tersteegen
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Clemens Cammann
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Eylin Topfstedt
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany
| | - Daniela Dieterich
- Institute of Pharmacology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Ulrike Seifert
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
22
|
Aygün C, Mutlu Ö. Computational characterisation of Toxoplasma gondii FabG (3-oxoacyl-[acyl-carrier-protein] reductase): a combined virtual screening and all-atom molecular dynamics simulation study. J Biomol Struct Dyn 2020; 40:1952-1969. [PMID: 33063633 DOI: 10.1080/07391102.2020.1834456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii is an opportunistic obligate parasite, ubiquitous around the globe with seropositivity rates that range from 10% to 90% and infection by the parasite of pregnant women causes pre-natal death of the foetus in most cases and severe neurodegenerative syndromes in some. No vaccine is currently available, and since drug-resistance is common among T. gondii strains, discovering lead compounds for drug design using diverse tactics is necessary. In this study, the sole constituent isoform of an enzymatic 3-oxoacyl-[acyl-carrier-protein] reduction step in an apicoplast-located fatty acid biosynthesis pathway was chosen as a possible drug target. FASII is prokaryotic therefore, targeting it would pose fewer side-effects to human hosts. After a homology 3D modelling of TgFabG, a high-throughput virtual screening of 9867 compounds, the elimination of ligands was carried out by a flexible ligand molecular docking and 200 ns molecular dynamics simulations, with additional DCCM and PC plot analyses. Molecular Dynamics and related post-MD analyses of the top 3 TgFabG binders selected for optimal free binding energies, showed that L2 maintained strong H-bonds with TgFabG and facilitated structural reorientation expected of FabGs, namely an expansion of the Rossmann Fold and a flexible lid capping. The most flexible TgFabG sites were the α7 helix (the flexible lid region) and the β4-α4 and β5-α6 loops. For TgFabG-L2, the movements of these regions toward the active site enabled greater ligand stability. Thus, L2 ("Skimmine"; PubChem ID: 320361), was ultimately selected as the optimal candidate for the discovery of lead compounds for rational drug design.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Can Aygün
- Faculty of Arts and Sciences, Biology Department, Marmara University, Istanbul, Turkey
| | - Özal Mutlu
- Faculty of Arts and Sciences, Biology Department, Marmara University, Istanbul, Turkey
| |
Collapse
|
23
|
Kong L, Jiang D, He C, Xia J, Wei H, Zhou L, Peng H. TgROP18 targets IL20RB for host-defense-related-STAT3 activation during Toxoplasma gondii infection. Parasit Vectors 2020; 13:400. [PMID: 32767999 PMCID: PMC7412674 DOI: 10.1186/s13071-020-04251-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/20/2020] [Indexed: 11/17/2022] Open
Abstract
Background Toxoplasma gondii is an opportunistic protozoan infecting almost one-third of the world’s population. Toxoplasma gondii rhoptry protein 18 (TgROP18) is a key virulence factor determining the parasite’s acute virulence and is secreted into host cells during infection. We previously identified the interaction of TgROP18 and host cell immune-related receptor protein IL20RB, and observed the activation of STAT3 in human keratinocytes (HaCaT) cells infected by the rop16 knockout RH strain, though TgROP16 is regarded as being responsible for host STAT3 activation during T. gondii invasion. Therefore, we hypothesize TgROP18 can activate host STAT3 through binding to IL20RB. Methods CRISPR-CAS9 technology was used to generate the ROP16 and ROP18 double knockout RH strain, RH-∆rop16∆rop18. SDS-PAGE and western blot were used to detect STAT3 activation in different HaCaT cells with high endogenous IL20RB expression treated with T. gondii tachyzoites infection, recombinant ROP18, or IL-20. FRET and co-immunoprecipitation (Co-IP) was used to detect the protein-protein interaction. Results We observed that TgROP18 was involved in a synergic activation of the host JAK/STAT3 pathway together with TgROP16 in human HaCaT cells infected with T. gondii or treated with recombinant TgROP18 protein, stimulating host proinflammatory immune responses such as expression of TNF-α. The effect of recombinant ROP18 on STAT3 phosphorylation was presented in a dose-dependent manner. Additionally, TgROP18 was identified to target IL20RB on its extracellular domain. When we treated different cell lines with the recombinant ROP18, STAT3 phosphorylation could only be observed in the cells with endogenous IL20RB expression, such as HaCaT cells. Conclusions These findings indicate that TgROP18-IL20RB interaction upon T. gondii invasion was involved in STAT3 activation, which is associated with host cell defense.![]()
Collapse
Affiliation(s)
- Ling Kong
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Dan Jiang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Cheng He
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Jing Xia
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Haixia Wei
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Lijuan Zhou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|
24
|
Azimi-Resketi M, Eskandarian A, Ganjalikhani-Hakemi M, Zohrabi T. Knocking down of the DHFR-TS gene in Toxoplasma gondii using siRNA and assessing the subsequences on toxoplasmosis in mice. Acta Trop 2020; 207:105488. [PMID: 32277926 DOI: 10.1016/j.actatropica.2020.105488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 02/05/2023]
Abstract
Toxoplasma gondii (T. gondii), an obligatory intracellular parasite, is the etiologic agent of toxoplasmosis. Dihydrofolate reductase-thymidylate synthase (DHFR-TS) is one of the most important enzymes in toxoplasma folic acid cycle. Due to the emergence of resistance in RH strain of T. gondii against pyrimethamine that acts via DHFR-TS inhibition and also the crucial role of small interference RNA (siRNA) technology in gene silencing, we aimed to use siRNA to knock down DHFR-TS gene expression in T. gondii as a therapeutic target against toxoplasmosis in a mouse model. Based on the DHFR-TS gene sequence, siRNA was designed. The siRNAs were transfected into the parasites by electroporation. Total RNA was extracted using RNX-Plus kit. The viability of parasite was assessed by methylthiazole tetrazolium (MTT). The survival time of mice challenged with siRNA-treated T.gondii were compared to the control group infected with the same amount of wild-type tachyzoites. The viability of siRNA-embedded parasites was 70.7% (29.3% decreased) compared to the wild-type parasite as control (P = 0.0001). The transcription level of siRNA-transfected parasites was reduced to 17.4% (82.6% inhibition) (P = 0.016). The in vivo assessment showed that the mean survival time of the mice inoculated with modified parasites was increased about 2 days after the death of all mice in the control group. The designed siRNAs in the current study were able to silence the DHFR-TS gene efficiently. This silencing led to a decrease in viability of the parasites and an increase in the survival time of the parasites-treated mice.
Collapse
Affiliation(s)
- Mojtaba Azimi-Resketi
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbasali Eskandarian
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mazdak Ganjalikhani-Hakemi
- Department of Medical Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | | |
Collapse
|
25
|
Ma Z, Yan K, Jiang R, Guan J, Yang L, Huang Y, Lu B, Li X, Zhang J, Chang Y, Wu X. A Novel wx2 Gene of Toxoplasma gondii Inhibits the Parasitic Invasion and Proliferation in vitro and Attenuates Virulence in vivo via Immune Response Modulation. Front Microbiol 2020; 11:399. [PMID: 32318029 PMCID: PMC7154108 DOI: 10.3389/fmicb.2020.00399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/26/2020] [Indexed: 11/25/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular apicomplexan protozoan that can parasitize most warm-blooded animals and cause severe diseases in immunocompromised individuals or fetal abnormalities in pregnant woman. The treatment of toxoplamosis has been limited by effective drugs. Our previous work indicated that the novel gene wx2 of T. gondii may serve as a vaccine antigen candidate. To further investigate the molecular functions of wx2 in highly virulent T. gondii (RH strain), a wx2 gene deletion mutant RH strain (KO-wx2) was established using CRISPR-Cas9. The phynotype of KO-wx2 was analyzed by plaque, invasion, and replication assays in vitro as well as in vivo virulence assays. The results indicated that the targeted deletion of the wx2 gene significantly inhibited in vitro parasite growth and replication in the host cells as well as attenuated parasite virulence in the mouse model. Notably, the percentage of pro-inflammatory factors of interferon gamma (IFN-γ) and interlukin-17A (IL-17A) and anti-inflammatory factor of interlukin-10 (IL-10) in the lymph nodes were upregulated in mice infected with the KO-wx2 strain. Our data suggested that the wx2 gene plays an important role in the process of the parasite’s life cycle and virulence in mice. In addition, it also plays an important role in the host’s immunity reaction, mainly via Th1 and Th17 cellular immunity, not Th2.
Collapse
Affiliation(s)
- Zhenrong Ma
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Kang Yan
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Ruolan Jiang
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Jie Guan
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Linfei Yang
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Yehong Huang
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Bin Lu
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Xuanwu Li
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Jie Zhang
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Yunfeng Chang
- Department of Forensic Medicine Science, Central South University, Changsha, China
| | - Xiang Wu
- Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| |
Collapse
|
26
|
Kano SI, Hodgkinson CA, Jones-Brando L, Eastwood S, Ishizuka K, Niwa M, Choi EY, Chang DJ, Chen Y, Velivela SD, Leister F, Wood J, Chowdari K, Ducci F, Caycedo DA, Heinz E, Newman ER, Cascella N, Mortensen PB, Zandi PP, Dickerson F, Nimgaonkar V, Goldman D, Harrison PJ, Yolken RH, Sawa A. Host-parasite interaction associated with major mental illness. Mol Psychiatry 2020; 25:194-205. [PMID: 30127472 PMCID: PMC6382596 DOI: 10.1038/s41380-018-0217-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/16/2018] [Accepted: 06/20/2018] [Indexed: 11/23/2022]
Abstract
Clinical studies frequently report that patients with major mental illness such as schizophrenia and bipolar disorder have co-morbid physical conditions, suggesting that systemic alterations affecting both brain and peripheral tissues might underlie the disorders. Numerous studies have reported elevated levels of anti-Toxoplasma gondii (T. gondii) antibodies in patients with major mental illnesses, but the underlying mechanism was unclear. Using multidisciplinary epidemiological, cell biological, and gene expression profiling approaches, we report here multiple lines of evidence suggesting that a major mental illness-related susceptibility factor, Disrupted in schizophrenia (DISC1), is involved in host immune responses against T. gondii infection. Specifically, our cell biology and gene expression studies have revealed that DISC1 Leu607Phe variation, which changes DISC1 interaction with activating transcription factor 4 (ATF4), modifies gene expression patterns upon T. gondii infection. Our epidemiological data have also shown that DISC1 607 Phe/Phe genotype was associated with higher T. gondii antibody levels in sera. Although further studies are required, our study provides mechanistic insight into one of the few well-replicated serological observations in major mental illness.
Collapse
Affiliation(s)
- Shin-Ichi Kano
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Colin A Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Lorraine Jones-Brando
- Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sharon Eastwood
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, United Kingdom
| | - Koko Ishizuka
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Minae Niwa
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Eric Y Choi
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Daniel J Chang
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Yian Chen
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Swetha D Velivela
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Flora Leister
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Joel Wood
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kodavali Chowdari
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Francesca Ducci
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Daniel A Caycedo
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Elizabeth Heinz
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Emily R Newman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Nicola Cascella
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Preben B Mortensen
- National Centre for Register-Based Research, University of Aarhus, Aarhus, 8000, Denmark
| | - Peter P Zandi
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, 21204, USA
| | - Vishwajit Nimgaonkar
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, United Kingdom
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Akira Sawa
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
27
|
Zhao N, Ming S, Lu Y, Wang F, Li H, Zhang X, Zhao X. Identification and Application of Epitopes in EtMIC1 of Eimeria tenella Recognized by the Monoclonal Antibodies 1-A1 and 1-H2. Infect Immun 2019; 87:e00596-19. [PMID: 31427452 PMCID: PMC6803336 DOI: 10.1128/iai.00596-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
Eimeria tenella microneme-1 protein (EtMIC1) has been proposed to be a transmembrane protein, but this characteristic has not yet been confirmed experimentally. Furthermore, despite EtMIC1 being an important candidate antigen, its key epitope has not been reported. Here, two linear B-cell epitopes of EtMIC1, 91LITFATRSK99 and 698ESLISAGE705, were identified by Western blotting using specific monoclonal antibodies (MAbs) and were named epitope I (located in the I-domain) and epitope CTR (located in the CTR domain), respectively. Sequence comparative analyses of these epitopes among Eimeria species that infect chickens showed that epitope I differs greatly across species, whereas epitope CTR is relatively conserved. Point mutation assay results indicate that all the amino acid residues of the epitopes recognized by MAb 1-A1 or 1-H2 are key amino acids involved in recognition. Comparative analyses of indirect immunofluorescence assay (IFA) results for MAbs 1-A1 and 1-H2 under both nonpermeabilization and permeabilization conditions indicate that epitope I is located on the outer side of the sporozoite surface membrane whereas epitope CTR is located on the inner side, together providing experimental evidence that EtMIC1 is a transmembrane protein. IFA also labeled the EtMIC1 protein on the parasitophorous vacuole membrane and on the surface of schizonts, which suggests that the EtMIC1 protein may play an important role in parasitophorous vacuole formation and E. tenella development. Immunoprotective efficacy experiments revealed that epitope I has good immunogenicity, as evidenced by its induction of high serum antibody levels, blood lymphocyte proliferation, and CD4+ blood lymphocyte percentage.
Collapse
Affiliation(s)
- Ningning Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Shuzhen Ming
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Yaru Lu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Fangkun Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Hongmei Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Xiao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Xiaomin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China
| |
Collapse
|
28
|
Thornton LB, Teehan P, Floyd K, Cochrane C, Bergmann A, Riegel B, Stasic AJ, Di Cristina M, Moreno SNJ, Roepe PD, Dou Z. An ortholog of Plasmodium falciparum chloroquine resistance transporter (PfCRT) plays a key role in maintaining the integrity of the endolysosomal system in Toxoplasma gondii to facilitate host invasion. PLoS Pathog 2019; 15:e1007775. [PMID: 31170269 PMCID: PMC6553793 DOI: 10.1371/journal.ppat.1007775] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 04/22/2019] [Indexed: 12/13/2022] Open
Abstract
Toxoplasma gondii is an apicomplexan parasite with the ability to use foodborne, zoonotic, and congenital routes of transmission that causes severe disease in immunocompromised patients. The parasites harbor a lysosome-like organelle, termed the "Vacuolar Compartment/Plant-Like Vacuole" (VAC/PLV), which plays an important role in maintaining the lytic cycle and virulence of T. gondii. The VAC supplies proteolytic enzymes that contribute to the maturation of invasion effectors and that digest autophagosomes and endocytosed host proteins. Previous work identified a T. gondii ortholog of the Plasmodium falciparum chloroquine resistance transporter (PfCRT) that localized to the VAC. Here, we show that TgCRT is a membrane transporter that is functionally similar to PfCRT. We also genetically ablate TgCRT and reveal that the TgCRT protein plays a key role in maintaining the integrity of the parasite’s endolysosomal system by controlling morphology of the VAC. When TgCRT is absent, the VAC dramatically increases in volume by ~15-fold and overlaps with adjacent endosome-like compartments. Presumably to reduce aberrant swelling, transcription and translation of endolysosomal proteases are decreased in ΔTgCRT parasites. Expression of subtilisin protease 1 is significantly reduced, which impedes trimming of microneme proteins, and significantly decreases parasite invasion. Chemical or genetic inhibition of proteolysis within the VAC reverses these effects, reducing VAC size and partially restoring integrity of the endolysosomal system, microneme protein trimming, and invasion. Taken together, these findings reveal for the first time a physiological role of TgCRT in substrate transport that impacts VAC volume and the integrity of the endolysosomal system in T. gondii. Toxoplasma gondii is an obligate intracellular protozoan parasite that belongs to the phylum Apicomplexa and that infects virtually all warm-blooded organisms. Approximately one-third of the human population is infected with Toxoplasma. Toxoplasma invades host cells using processed invasion effectors. A lysosome-like organelle (VAC) is involved in refining these invasion effectors to reach their final forms. A T. gondii ortholog of the malarial chloroquine resistance transporter protein (TgCRT) was found to be localized to the VAC membrane. Although the mutated version of the malarial chloroquine resistance transporter (PfCRT) has been shown to confer resistance to chloroquine treatment, its physiologic function remains poorly understood. Comparison between the related PfCRT and TgCRT facilitates definition of the physiologic role of CRT proteins. Here, we report that TgCRT plays a key role in affecting the integrity and proteolytic activity of the VAC and adjacent organelles, the secretion of invasion effectors, and parasite invasion and virulence. To relieve osmotic stress caused by VAC swelling when TgCRT is deleted, parasites repress proteolysis within this organelle to decrease solute accumulation, which then has secondary effects on parasite invasion. Our findings highlight a common function for PfCRT and TgCRT in mediating small solute transport to affect apicomplexan parasite vacuolar size and function.
Collapse
Affiliation(s)
- L. Brock Thornton
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Paige Teehan
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Katherine Floyd
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Christian Cochrane
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Amy Bergmann
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Bryce Riegel
- Department of Chemistry, Georgetown University, NW, Washington DC, United States of America
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, NW, Washington DC, United States of America
| | - Andrew J. Stasic
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Manlio Di Cristina
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Silvia N. J. Moreno
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Paul D. Roepe
- Department of Chemistry, Georgetown University, NW, Washington DC, United States of America
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, NW, Washington DC, United States of America
| | - Zhicheng Dou
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
29
|
Pereira ACA, Silva RJ, Franco PS, de Oliveira Gomes A, Souza G, Milian ICB, Ribeiro M, Rosini AM, Guirelli PM, Ramos ELP, Mineo TWP, Mineo JR, Silva NM, Ferro EAV, Barbosa BF. Cyclooxygenase (COX)-2 Inhibitors Reduce Toxoplasma gondii Infection and Upregulate the Pro-inflammatory Immune Response in Calomys callosus Rodents and Human Monocyte Cell Line. Front Microbiol 2019; 10:225. [PMID: 30809216 PMCID: PMC6379304 DOI: 10.3389/fmicb.2019.00225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/28/2019] [Indexed: 12/27/2022] Open
Abstract
Toxoplasma gondii is able to infect a wide range of vertebrates, including humans. Studies show that cyclooxygenase-2 (COX-2) is a modulator of immune response in multiple types of infection, such as Trypanosoma cruzi. However, the role of COX-2 during T. gondii infection is still unclear. The aim of this study was to investigate the role of COX-2 during infection by moderately or highly virulent strains of T. gondii in Calomys callosus rodents and human THP-1 cells. C. callosus were infected with 50 cysts of T. gondii (ME49), treated with COX-2 inhibitors (meloxicam or celecoxib) and evaluated to check body weight and morbidity. After 40 days, brain and serum were collected for detection of T. gondii by real-time PCR and immunohistochemistry or cytokines by CBA. Furthermore, peritoneal macrophages or THP-1 cells, infected with RH strain or uninfected, were treated with meloxicam or celecoxib to evaluate the parasite proliferation by colorimetric assay and cytokine production by ELISA. Finally, in order to verify the role of prostaglandin E2 in COX-2 mechanism, THP-1 cells were infected, treated with meloxicam or celecoxib plus PGE2, and analyzed to parasite proliferation and cytokine production. The data showed that body weight and morbidity of the animals changed after infection by T. gondii, under both treatments. Immunohistochemistry and real-time PCR showed a reduction of T. gondii in brains of animals treated with both COX-2 inhibitors. Additionally, it was observed that both COX-2 inhibitors controlled the T. gondii proliferation in peritoneal macrophages and THP-1 cells, and the treatment with PGE2 restored the parasite growth in THP-1 cells blocked to COX-2. In the serum of Calomys, upregulation of pro-inflammatory cytokines was detected, while the supernatants of peritoneal macrophages and THP-1 cells demonstrated significant production of TNF and nitrite, or TNF, nitrite and MIF, respectively, under both COX-2 inhibitors. Finally, PGE2 treatment in THP-1 cells triggered downmodulation of pro-inflammatory mediators and upregulation of IL-8 and IL-10. Thus, COX-2 is an immune mediator involved in the susceptibility to T. gondii regardless of strain or cell types, since inhibition of this enzyme induced control of infection by upregulating important pro-inflammatory mediators against Toxoplasma.
Collapse
Affiliation(s)
- Ana Carolina Alcântara Pereira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Angelica de Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Guilherme Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Iliana Claudia Balga Milian
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Pâmela Mendonça Guirelli
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eliézer Lucas Pires Ramos
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| |
Collapse
|
30
|
Li JX, He JJ, Elsheikha HM, Chen D, Zhai BT, Zhu XQ, Yan HK. Toxoplasma gondii ROP17 inhibits the innate immune response of HEK293T cells to promote its survival. Parasitol Res 2019; 118:783-792. [PMID: 30675671 DOI: 10.1007/s00436-019-06215-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/10/2019] [Indexed: 10/27/2022]
Abstract
Toxoplasma gondii secretes a group of rhoptry-secreted kinases (ROPs), which play significant roles in promoting intracellular infection. T. gondii rhoptry organelle protein 17 (ROP17) is one of these important effector proteins. However, its role in modulating host cell response during infection remains poorly understood. Here, we reveal that ROP17 (genotype I) induces significant changes in the expression genes and transcription factors of host cells. HEK293T cells were transfected with PCMV-N-HA-ROP17 plasmid or empty control PCMV-N-HA plasmid. Transcriptomic analysis revealed 3138 differentially expressed genes (DEGs) in PCMV-N-HA-ROP17-transfected HEK293T cells, including 1456 upregulated, 1682 downregulated DEGs. Also, 715 of the DEGs were transcription factors (TFs), including 423 downregulated TFs and 292 upregulated TFs. Most differentially expressed TFs, whether belong to signal transduction, cancer-related pathways or immune-related pathways, were downregulated in ROP17-expressing cells. ROP17 also decreased alternative splicing events in host cells, presumably via alteration of the expression of genes involved in the alternative splicing pathway. Taken together, our findings suggest a novel strategy whereby T. gondii ROP17 manipulates various cellular processes, including immune response through reprogramming host gene expression to promote its own colonization and survival in the infected host cells.
Collapse
Affiliation(s)
- Jie-Xi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong Province, People's Republic of China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Dan Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China
| | - Bin-Tao Zhai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China.,Inner Mongolia Agriculture University, Hohhot, 010018, People's Republic of China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China. .,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University College of Veterinary Medicine, Yangzhou, 225009, Jiangsu Province, People's Republic of China.
| | - Hai-Kuo Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong Province, People's Republic of China.
| |
Collapse
|
31
|
Lauri N, Bazzi Z, Alvarez CL, Leal Denis MF, Schachter J, Herlax V, Ostuni MA, Schwarzbaum PJ. ATPe Dynamics in Protozoan Parasites. Adapt or Perish. Genes (Basel) 2018; 10:E16. [PMID: 30591699 PMCID: PMC6356682 DOI: 10.3390/genes10010016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 01/25/2023] Open
Abstract
In most animals, transient increases of extracellular ATP (ATPe) are used for physiological signaling or as a danger signal in pathological conditions. ATPe dynamics are controlled by ATP release from viable cells and cell lysis, ATPe degradation and interconversion by ecto-nucleotidases, and interaction of ATPe and byproducts with cell surface purinergic receptors and purine salvage mechanisms. Infection by protozoan parasites may alter at least one of the mechanisms controlling ATPe concentration. Protozoan parasites display their own set of proteins directly altering ATPe dynamics, or control the activity of host proteins. Parasite dependent activation of ATPe conduits of the host may promote infection and systemic responses that are beneficial or detrimental to the parasite. For instance, activation of organic solute permeability at the host membrane can support the elevated metabolism of the parasite. On the other hand ecto-nucleotidases of protozoan parasites, by promoting ATPe degradation and purine/pyrimidine salvage, may be involved in parasite growth, infectivity, and virulence. In this review, we will describe the complex dynamics of ATPe regulation in the context of protozoan parasite⁻host interactions. Particular focus will be given to features of parasite membrane proteins strongly controlling ATPe dynamics. This includes evolutionary, genetic and cellular mechanisms, as well as structural-functional relationships.
Collapse
Affiliation(s)
- Natalia Lauri
- Institute of Biological Chemistry and Physicochemistry (IQUIFIB) "Prof. Alejandro C. Paladini", Faculty of Pharmacy and Biochemistry, University of Buenos Aires, National Scientific and Technical Research Council (CONICET), Junín 956 Buenos Aires, Argentina.
- Faculty of Pharmacy and Biochemistry, Department of Biological Chemistry, Chair of Biological Chemistry, University of Buenos Aires, Junín 956 Buenos Aires, Argentina.
| | - Zaher Bazzi
- Institute of Biological Chemistry and Physicochemistry (IQUIFIB) "Prof. Alejandro C. Paladini", Faculty of Pharmacy and Biochemistry, University of Buenos Aires, National Scientific and Technical Research Council (CONICET), Junín 956 Buenos Aires, Argentina.
| | - Cora L Alvarez
- Institute of Biological Chemistry and Physicochemistry (IQUIFIB) "Prof. Alejandro C. Paladini", Faculty of Pharmacy and Biochemistry, University of Buenos Aires, National Scientific and Technical Research Council (CONICET), Junín 956 Buenos Aires, Argentina.
- Faculty of Exact and Natural Sciences, Department of Biodiversity and Experimental Biology, University of Buenos Aires, Intendente Güiraldes, Buenos Aires 2160, Argentina.
| | - María F Leal Denis
- Institute of Biological Chemistry and Physicochemistry (IQUIFIB) "Prof. Alejandro C. Paladini", Faculty of Pharmacy and Biochemistry, University of Buenos Aires, National Scientific and Technical Research Council (CONICET), Junín 956 Buenos Aires, Argentina.
- Chair of Analytical Chemistry and Physicochemistry, Faculty of Pharmacy and Biochemistry, Department of Analytical Chemistry, University of Buenos Aires, Junín 956 Buenos Aires, Argentina.
| | - Julieta Schachter
- Institute of Biological Chemistry and Physicochemistry (IQUIFIB) "Prof. Alejandro C. Paladini", Faculty of Pharmacy and Biochemistry, University of Buenos Aires, National Scientific and Technical Research Council (CONICET), Junín 956 Buenos Aires, Argentina.
| | - Vanesa Herlax
- Biochemistry Research Institute of La Plata (INIBIOLP) "Prof. Dr. Rodolfo R. Brenner", Faculty of Medical Sciences, National University of La Plata, National Scientific and Technical Research Council, Av. 60 y Av. 120 La Plata, Argentina.
- National University of La Plata, Faculty of Medical Sciences, Av. 60 y Av. 120 La Plata, Argentina.
| | - Mariano A Ostuni
- UMR-S1134, Integrated Biology of Red Blood Cells, INSERM, Paris Diderot University, Sorbonne Paris Cité, University of La Réunion, University of Antilles, F-75015 Paris, France.
- National Institute of Blood Transfusion (INTS), Laboratory of Excellence GR-Ex, F-75015 Paris, France.
| | - Pablo J Schwarzbaum
- Institute of Biological Chemistry and Physicochemistry (IQUIFIB) "Prof. Alejandro C. Paladini", Faculty of Pharmacy and Biochemistry, University of Buenos Aires, National Scientific and Technical Research Council (CONICET), Junín 956 Buenos Aires, Argentina.
- Faculty of Pharmacy and Biochemistry, Department of Biological Chemistry, Chair of Biological Chemistry, University of Buenos Aires, Junín 956 Buenos Aires, Argentina.
| |
Collapse
|
32
|
He C, Kong L, Puthiyakunnon S, Wei HX, Zhou LJ, Peng HJ. iTRAQ-based phosphoproteomic analysis reveals host cell's specific responses to Toxoplasma gondii at the phases of invasion and prior to egress. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1867:202-212. [PMID: 30576742 DOI: 10.1016/j.bbapap.2018.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 10/27/2022]
Abstract
Protein phosphorylation plays a key role in host cell-T. gondii interaction. However, the phosphoproteome data of host cell at various phases of T. gondii infection has not been thoroughly described. In this study, we assessed the host phosphoproteome data with isobaric tags for relative and absolute quantification (iTRAQ) method during the phases of T. gondii invasion (30 min post infection, PI) and prior to egress (28 h PI). Our iTRAQ analysis revealed a total of 665 phosphoproteins, among which the significantly regulated phosphoproteins in different between-group comparisons were further analyzed. Functional analysis of these significantly regulated phosphoproteins suggested that T. gondii modulated host cell processes through phosphorylation including cell cycle regulation, inducing apoptosis, blocking the synthesis of some inflammatory factors, mediating metabolism to support its proliferation at the infection phase prior to egress, and utilizing membrane and energy from host cell, reorganizing cytoskeleton to favor its invasion and PV formation at the phase of invasion. The phosphorylation level of Smad2, CTNNA1, and HSPB1 identified with western blot revealed a consistent trend of change with iTRAQ result. These newly identified and significantly regulated phosphoproteins from our phosphoproteome data may provide new clues to unravel the host cell's complex reaction against T. gondii infection and the interaction between the host cell and T. gondii.
Collapse
Affiliation(s)
- Cheng He
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Ling Kong
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Santhosh Puthiyakunnon
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hai-Xia Wei
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Li-Juan Zhou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hong-Juan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
33
|
Danielson JJ, Perez N, Romano JD, Coppens I. Modelling Toxoplasma gondii infection in a 3D cell culture system In Vitro: Comparison with infection in 2D cell monolayers. PLoS One 2018; 13:e0208558. [PMID: 30521607 PMCID: PMC6283583 DOI: 10.1371/journal.pone.0208558] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional (3D) cell culture models bridge the gap between two-dimensional (2D) monolayer cultures and animal models. Physiologically relevant, 3D culture models have significantly advanced basic cell science and provide unique insights into host-pathogen interactions intrinsically linked to cell morphology. Toxoplasma gondii is an obligate intravacuolar parasite that chronically infects a large portion of the global human population. Our current understanding of Toxoplasma infection is largely based on 2D cell cultures, in which mammalian cells are grown on flat surfaces. However, 2D cell cultures may not recapitulate key conditions of in vivo infections as they introduce artificial pressures and tensions, which may subsequently alter infectious processes that are dependent on spatiality, e.g., invasion, replication and egress. In this study, we adapted a collagen-based 3D cell culture system to reproduce the 3D environment of T. gondii natural infections for investigation of the replication and egress of the parasite from the parasitophorous vacuole. Suspended in the 3D matrix, Toxoplasma-infected VERO cells have round morphology, as opposed to infected VERO cells in 2D monolayers. The doubling time of Toxoplasma in VERO cells within the matrix is comparable to that of parasites cultivated in VERO cell monolayers. In the absence of the pressure of flattened host cells grown in 2D cultures, the parasitophorous vacuole of T. gondii has a globular shape, with intravacuolar parasites distributed radially, forming 3D spherical ‘rosette’ structures. Parasites egress radially away from the ruptured host cell in 3D matrices, in contrast to Toxoplasma cultivated in 2D monolayer cultures, where the parasites escape perpendicularly from the flat surface below the host cells. These observations demonstrate the utility of collagen matrices for studying parasite modes of infection as these 3D assays more closely mimic in vivo conditions.
Collapse
Affiliation(s)
- Jeffrey J. Danielson
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Nicolas Perez
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
34
|
Rocha-Roa C, Molina D, Cardona N. A Perspective on Thiazolidinone Scaffold Development as a New Therapeutic Strategy for Toxoplasmosis. Front Cell Infect Microbiol 2018; 8:360. [PMID: 30386743 PMCID: PMC6198644 DOI: 10.3389/fcimb.2018.00360] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/26/2018] [Indexed: 11/17/2022] Open
Abstract
Toxoplasma gondii is one of the most successful parasites due to its ability to infect a wide variety of warm-blooded animals. It is estimated that one-third of the world's population is latently infected. The generic therapy for toxoplasmosis has been a combination of antifolates such as pyrimethamine or trimethoprim with either sulfadiazine or antibiotics such as clindamycin with a combination with leucovorin to prevent hematologic toxicity. This therapy shows limitations such as drug intolerance, low bioavailability or drug resistance by the parasite. There is a need for the development of new molecules with the capacity to block any stage of the parasite's life cycle in humans or in a different type of hosts. Heterocyclic compounds are promissory drugs due to its reported biological activity; for this reason, thiazolidinone and its derivatives are presented as a new alternative not only for its inhibitory activity against the parasite but also for its high selectivity-level with high therapeutic index. Thiazolidinones are an important scaffold known to be associated with anticancer, antibacterial, antifungal, antiviral, antioxidant, and antidiabetic activities. The molecule possesses an imidazole ring that has been described as an antiprotozoal agent with antiparasitic properties and less toxicity. Thiazolidinone derivatives have been reportedly as building blocks in organic chemistry and as scaffolds for drug discovery. Here we present a perspective of how structural modifications of the thiazolidinone core could generate new compounds with high anti-parasitic effect and less toxic results.
Collapse
Affiliation(s)
- Cristian Rocha-Roa
- Centre for Biomedical Research CIBM, University of Quindío, Armenia, Colombia
| | - Diego Molina
- Centre for Biomedical Research CIBM, University of Quindío, Armenia, Colombia
| | - Néstor Cardona
- Centre for Biomedical Research CIBM, University of Quindío, Armenia, Colombia.,Dentistry Faculty, University Antonio Nariño, Armenia, Colombia
| |
Collapse
|
35
|
Farhat S, Florent I, Noel B, Kayal E, Da Silva C, Bigeard E, Alberti A, Labadie K, Corre E, Aury JM, Rombauts S, Wincker P, Guillou L, Porcel BM. Comparative Time-Scale Gene Expression Analysis Highlights the Infection Processes of Two Amoebophrya Strains. Front Microbiol 2018; 9:2251. [PMID: 30333799 PMCID: PMC6176090 DOI: 10.3389/fmicb.2018.02251] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/04/2018] [Indexed: 01/31/2023] Open
Abstract
Understanding factors that generate, maintain, and constrain host-parasite associations is of major interest to biologists. Although little studied, many extremely virulent micro-eukaryotic parasites infecting microalgae have been reported in the marine plankton. This is the case for Amoebophrya, a diverse and highly widespread group of Syndiniales infecting and potentially controlling dinoflagellate populations. Here, we analyzed the time-scale gene expression of a complete infection cycle of two Amoebophrya strains infecting the same host (the dinoflagellate Scrippsiella acuminata), but diverging by their host range (one infecting a single host, the other infecting more than one species). Over two-thirds of genes showed two-fold differences in expression between at least two sampled stages of the Amoebophrya life cycle. Genes related to carbohydrate metabolism as well as signaling pathways involving proteases and transporters were overexpressed during the free-living stage of the parasitoid. Once inside the host, all genes related to transcription and translation pathways were actively expressed, suggesting the rapid and extensive protein translation needed following host-cell invasion. Finally, genes related to cellular division and components of the flagellum organization were overexpressed during the sporont stage. In order to gain a deeper understanding of the biological basis of the host-parasitoid interaction, we screened proteins involved in host-cell recognition, invasion, and protection against host-defense identified in model apicomplexan parasites. Very few of the genes encoding critical components of the parasitic lifestyle of apicomplexans could be unambiguously identified as highly expressed in Amoebophrya. Genes related to the oxidative stress response were identified as highly expressed in both parasitoid strains. Among them, the correlated expression of superoxide dismutase/ascorbate peroxidase in the specialist parasite was consistent with previous studies on Perkinsus marinus defense. However, this defense process could not be identified in the generalist Amoebophrya strain, suggesting the establishment of different strategies for parasite protection related to host specificity.
Collapse
Affiliation(s)
- Sarah Farhat
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ. Evry, Université Paris-Saclay, Evry, France
| | - Isabelle Florent
- Communication Molecules and Adaptation of Microorganisms, National Museum of Natural History, CNRS, Paris, France
| | - Benjamin Noel
- Genoscope, Institut François Jacob, CEA, Evry, France
| | - Ehsan Kayal
- Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, CNRS, UMR 7144, Station Biologique de Roscoff, Roscoff, France
| | | | - Estelle Bigeard
- Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, CNRS, UMR 7144, Station Biologique de Roscoff, Roscoff, France
| | | | | | - Erwan Corre
- Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, CNRS, UMR 7144, Station Biologique de Roscoff, Roscoff, France
| | | | - Stephane Rombauts
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Patrick Wincker
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ. Evry, Université Paris-Saclay, Evry, France
| | - Laure Guillou
- Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, CNRS, UMR 7144, Station Biologique de Roscoff, Roscoff, France
| | - Betina M Porcel
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ. Evry, Université Paris-Saclay, Evry, France
| |
Collapse
|
36
|
Venugopal K, Marion S. Secretory organelle trafficking in Toxoplasma gondii: A long story for a short travel. Int J Med Microbiol 2018; 308:751-760. [DOI: 10.1016/j.ijmm.2018.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/10/2018] [Accepted: 07/15/2018] [Indexed: 12/15/2022] Open
|
37
|
The Protozoan Parasite Toxoplasma gondii Selectively Reprograms the Host Cell Translatome. Infect Immun 2018; 86:IAI.00244-18. [PMID: 29967092 DOI: 10.1128/iai.00244-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
The intracellular parasite Toxoplasma gondii promotes infection by targeting multiple host cell processes; however, whether it modulates mRNA translation is currently unknown. Here, we show that infection of primary murine macrophages with type I or II T. gondii strains causes a profound perturbation of the host cell translatome. Notably, translation of transcripts encoding proteins involved in metabolic activity and components of the translation machinery was activated upon infection. In contrast, the translational efficiency of mRNAs related to immune cell activation and cytoskeleton/cytoplasm organization was largely suppressed. Mechanistically, T. gondii bolstered mechanistic target of rapamycin (mTOR) signaling to selectively activate the translation of mTOR-sensitive mRNAs, including those with a 5'-terminal oligopyrimidine (5' TOP) motif and those encoding mitochondrion-related proteins. Consistent with parasite modulation of host mTOR-sensitive translation to promote infection, inhibition of mTOR activity suppressed T. gondii replication. Thus, selective reprogramming of host mRNA translation represents an important subversion strategy during T. gondii infection.
Collapse
|
38
|
Choi WH, Lee IA. Evaluation of Anti- Toxoplasma gondii Effect of Ursolic Acid as a Novel Toxoplasmosis Inhibitor. Pharmaceuticals (Basel) 2018; 11:E43. [PMID: 29747388 PMCID: PMC6026977 DOI: 10.3390/ph11020043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
This study was carried out to evaluate the anti-parasitic effect of ursolic acid against Toxoplasma gondii (T. gondii) that induces toxoplasmosis, particularly in humans. The anti-parasitic effects of ursolic acid against T. gondii-infected cells and T. gondii were evaluated through different specific assays, including immunofluorescence staining and animal testing. Ursolic acid effectively inhibited the proliferation of T. gondii when compared with sulfadiazine, and consistently induced anti-T. gondii activity/effect. In particular, the formation of parasitophorous vacuole membrane (PVM) in host cells was markedly decreased after treating ursolic acid, which was effectively suppressed. Moreover, the survival rate of T. gondii was strongly inhibited in T. gondii group treated with ursolic acid, and then 50% inhibitory concentration (IC50) against T. gondii was measured as 94.62 μg/mL. The T. gondii-infected mice treated with ursolic acid indicated the same survival rates and activity as the normal group. These results demonstrate that ursolic acid causes anti-T. gondii action and effect by strongly blocking the proliferation of T. gondii through the direct and the selective T. gondii-inhibitory ability as well as increases the survival of T. gondii-infected mice. This study shows that ursolic acid has the potential to be used as a promising anti-T. gondii candidate substance for developing effective anti-parasitic drugs.
Collapse
Affiliation(s)
- Won Hyung Choi
- Marine Bio Research & Education Center, Kunsan National University, 558 Daehak-ro, Gunsan-si, Jeollabuk-do 54150, Korea.
| | - In Ah Lee
- Department of Chemistry, College of Natural Science, Kunsan National University, 558 Daehak-ro, Gunsan-si, Jeollabuk-do 54150, Korea.
| |
Collapse
|
39
|
Long-term affected flat oyster (Ostrea edulis) haemocytes show differential gene expression profiles from naïve oysters in response to Bonamia ostreae. Genomics 2018; 110:390-398. [PMID: 29678683 DOI: 10.1016/j.ygeno.2018.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/15/2018] [Accepted: 04/06/2018] [Indexed: 02/06/2023]
Abstract
European flat oyster (Ostrea edulis) production has suffered a severe decline due to bonamiosis. The responsible parasite enters in oyster haemocytes, causing an acute inflammatory response frequently leading to death. We used an immune-enriched oligo-microarray to understand the haemocyte response to Bonamia ostreae by comparing expression profiles between naïve (NS) and long-term affected (AS) populations along a time series (1 d, 30 d, 90 d). AS showed a much higher response just after challenge, which might be indicative of selection for resistance. No regulated genes were detected at 30 d in both populations while a notable reactivation was observed at 90 d, suggesting parasite latency during infection. Genes related to extracellular matrix and protease inhibitors, up-regulated in AS, and those related to histones, down-regulated in NS, might play an important role along the infection. Twenty-four candidate genes related to resistance should be further validated for selection programs aimed to control bonamiosis.
Collapse
|
40
|
Wei W, Zhang F, Chen H, Tang Y, Xing T, Luo Q, Yu L, Du J, Shen J, Zhang L. Toxoplasma gondii dense granule protein 15 induces apoptosis in choriocarcinoma JEG-3 cells through endoplasmic reticulum stress. Parasit Vectors 2018; 11:251. [PMID: 29665822 PMCID: PMC5904991 DOI: 10.1186/s13071-018-2835-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022] Open
Abstract
Background Toxoplasma gondii, a single-celled parasite commonly found in mammals, has been shown to induce trophoblast cell apoptosis and subsequently cause fetal damage and abortion. Although dense granule protein 15 (GRA15) has been identified as a key component in innate immunity to T. gondii infection and its pathogenesis, its role in host cell apoptosis remains unclarified. Methods Type II GRA15 (GRA15II) cDNA was inserted into a plasmid encoding enhanced green fluorescent protein (pEGFP). Choriocarcinoma JEG-3 cells were transfected with either pEGFP or pEGFP-GRA15II and cultured for 24 h. Cell apoptosis and endoplasmic reticulum stress (ERS) responses were assessed. Inhibitors targeting inositol-requiring kinase 1α (IRE1α; 4μ8C, 100 nM) or c-Jun N-terminal kinase (JNK; SP6000125, 20 μM) were added 12 h after plasmid transfection, followed by testing the effect of GRA15II on ERS. Results When compared to pEGFP, pEGFP-GRA15II transfection facilitated cell apoptosis (P < 0.05), increased mRNA expression of caspase-3, caspase-4, 78-kDa glucose-regulated protein (GRP78), C/EBP homologous protein (CHOP) and X-box binding protein-1 (XBP1) (all P < 0.05), and promoted protein expression of cleaved caspase-3, cleaved poly(ADP-ribose) polymerase, Bax, CHOP, GRP78, phospho-JNK, and phospho-IRE1α (all P < 0.05). The 4μ8C and SP6000125 decreased apoptosis and protein expression of XBP1s, CHOP, TNF receptor-associated factor 2 (TRAF2), phosphorylated apoptosis signal-regulating kinase 1 (ASK1), cleaved caspase-3, phospho-JNK, and Bax (all P < 0.05) in pEGFP-GRA15II transfected cells. Conclusions Toxoplasma GRA15II induced ERS and subsequently caused apoptosis of choriocarcinoma JEG-3 cells.
Collapse
Affiliation(s)
- Wei Wei
- Department of Immunology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Fangfang Zhang
- Department of Pathogen Biology and the Key Laboratory of Microbiology (Anhui), School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - He Chen
- Laboratory of Clinical Diagnostics, the First Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yuanyuan Tang
- Department of Pathogen Biology and the Key Laboratory of Microbiology (Anhui), School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Tian Xing
- Key Laboratory of Oral Diseases Research of Anhui Province, Hospital of Stomatology, Anhui Medical University, Hefei, 230032, China
| | - Qingli Luo
- Department of Pathogen Biology and the Key Laboratory of Microbiology (Anhui), School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Li Yu
- Department of Pathogen Biology and the Key Laboratory of Microbiology (Anhui), School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Jian Du
- Department of Pathogen Biology and the Key Laboratory of Microbiology (Anhui), School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Jilong Shen
- Department of Immunology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China. .,Department of Pathogen Biology and the Key Laboratory of Microbiology (Anhui), School of Basic Medicine, Anhui Medical University, Hefei, 230032, China. .,Laboratory of Clinical Diagnostics, the First Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Linjie Zhang
- Department of Immunology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
41
|
van der Linde K, Timofejeva L, Egger RL, Ilau B, Hammond R, Teng C, Meyers BC, Doehlemann G, Walbot V. Pathogen Trojan Horse Delivers Bioactive Host Protein to Alter Maize Anther Cell Behavior in Situ. THE PLANT CELL 2018; 30:528-542. [PMID: 29449414 PMCID: PMC5894838 DOI: 10.1105/tpc.17.00238] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 01/16/2018] [Accepted: 02/13/2018] [Indexed: 05/21/2023]
Abstract
Small proteins are crucial signals during development, host defense, and physiology. The highly spatiotemporal restricted functions of signaling proteins remain challenging to study in planta. The several month span required to assess transgene expression, particularly in flowers, combined with the uncertainties from transgene position effects and ubiquitous or overexpression, makes monitoring of spatiotemporally restricted signaling proteins lengthy and difficult. This situation could be rectified with a transient assay in which protein deployment is tightly controlled spatially and temporally in planta to assess protein functions, timing, and cellular targets as well as to facilitate rapid mutagenesis to define functional protein domains. In maize (Zea mays), secreted ZmMAC1 (MULTIPLE ARCHESPORIAL CELLS1) was proposed to trigger somatic niche formation during anther development by participating in a ligand-receptor module. Inspired by Homer's Trojan horse myth, we engineered a protein delivery system that exploits the secretory capabilities of the maize smut fungus Ustilago maydis, to allow protein delivery to individual cells in certain cell layers at precise time points. Pathogen-supplied ZmMAC1 cell-autonomously corrected both somatic cell division and differentiation defects in mutant Zmmac1-1 anthers. These results suggest that exploiting host-pathogen interactions may become a generally useful method for targeting host proteins to cell and tissue types to clarify cellular autonomy and to analyze steps in cell responses.
Collapse
Affiliation(s)
| | - Ljudmilla Timofejeva
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618 Tallinn, Estonia
| | - Rachel L Egger
- Department of Biology, Stanford University, Stanford, California 94305
| | - Birger Ilau
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618 Tallinn, Estonia
| | - Reza Hammond
- Donald Danforth Plant Science Center, St. Louis, Missouri 63132
| | - Chong Teng
- Donald Danforth Plant Science Center, St. Louis, Missouri 63132
| | - Blake C Meyers
- Donald Danforth Plant Science Center, St. Louis, Missouri 63132
| | - Gunther Doehlemann
- Botanical Institute and Cluster of Excellence on Plant Sciences (CEPLAS), University of Cologne, BioCenter, 50674 Cologne, Germany
| | - Virginia Walbot
- Department of Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
42
|
CHERAGHIPOUR K, SHARIATI L, KHANAHMAD H, GANJALIKHANI-HAKEMI M, MORIDNIA A, MIRIAN M, PESTEHCHIAN N. Induction of Apoptosis in Toxoplasma gondii Infected Hela Cells by Cisplatin and Sodium Azide and Isolation of Apoptotic Bodies and Potential Use for Vaccination against Toxoplasma gondii. IRANIAN JOURNAL OF PARASITOLOGY 2018; 13:406-415. [PMID: 30483332 PMCID: PMC6243158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Toxoplasma gondii can infect a wide range of mammalians, especially humans. It controls several intracellular signals for the inhibition of apoptosis. This study aimed to investigate the apoptogenic effect of cisplatin and sodium azide on T. gondii infected HeLa cells and isolate apoptotic bodies (blebs) as a potent stimulator of the immune system. METHODS The cytotoxic properties of cisplatin and sodium azide (NaN3) on HeLa cells were evaluated by MTT assay. Moreover, the apoptogenic activity of cisplatin and NaN3 was studied using flow cytometry (Annexin V/PI double staining) and scanning electron microscopy (SEM). Finally, apoptotic bodies were separated by centrifugation. RESULTS MTT assay data showed that the survival rate of cells treated with different concentration of NaN3 was significantly reduced, compared to negative control groups. Concerning cisplatin, only concentration of 20 μM had not a significant impact on the cell viability; however, the other concentration of cisplatin significantly reduced cell viability, compared to negative control groups. The level of early apoptosis in uninfected HeLa cells was higher compared to infected HeLa cells treated with cisplatin and NaN3. Finally, apoptotic bodies were separated from T. gondii infected HeLa cells treated with cisplatin. CONCLUSION Apoptosis was induced in both uninfected and infected HeLa cells with T. gondii and apoptotic bodies were isolated from infected cells. Therefore, further studies on apoptotic bodies are required in order to find a proper candidate for vaccine preparation against T. gondii infections.
Collapse
Affiliation(s)
- Kourosh CHERAGHIPOUR
- Dept. of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Laleh SHARIATI
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein KHANAHMAD
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Diseases, Isfahan University of Medical Sciences, Isfahan, Iran,Dept. of Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Abbas MORIDNIA
- Dept. of Immunology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Mina MIRIAN
- Dept. of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nader PESTEHCHIAN
- Dept. of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran,Correspondence
| |
Collapse
|
43
|
Roiko MS, LaFavers K, Leland D, Arrizabalaga G. Toxoplasma gondii-positive human sera recognise intracellular tachyzoites and bradyzoites with diverse patterns of immunoreactivity. Int J Parasitol 2017; 48:225-232. [PMID: 29170086 DOI: 10.1016/j.ijpara.2017.08.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 11/28/2022]
Abstract
Antibody detection assays have long been the first line test to confirm infection with the zoonotic parasite Toxoplasma gondii. However, challenges exist with serological diagnosis, especially distinguishing between acute, latent and reactivation disease states. The sensitivity and specificity of serological tests might be improved by testing for antibodies against parasite antigens other than those typically found on the parasite surface during the acute stage. To this end, we analysed the reactivity profile of human sera, identified as positive for anti-Toxoplasma gondii IgG in traditional assays, by indirect immunofluorescence reactivity to acute stage intracellular tachyzoites and in vitro-induced latent stage bradyzoites. The majority of anti-Toxoplasma gondii IgG positive sera recognised both intracellularly replicating tachyzoites and in vitro-induced bradyzoites with varying patterns of immune-reactivity. Furthermore, anti-bradyzoite antibodies were not detected in sera that were IgM-positive/IgG-negative. These results demonstrate that anti-Toxoplasma gondii-positive sera may contain antibodies to a variety of antigens in addition to those traditionally used in serological tests, and suggest the need for further investigations into the utility of anti-bradyzoite-specific antibodies to aid in diagnosis of Toxoplasma gondii infection.
Collapse
Affiliation(s)
- Marijo S Roiko
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, 350 West 11th Street, Indianapolis, IN, United States.
| | - Kaice LaFavers
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, MS A-519, Indianapolis, IN, United States
| | - Diane Leland
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, 350 West 11th Street, Indianapolis, IN, United States
| | - Gustavo Arrizabalaga
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 635 Barnhill Drive, MS A-519, Indianapolis, IN, United States
| |
Collapse
|
44
|
Chakraborty S, Roy S, Mistry HU, Murthy S, George N, Bhandari V, Sharma P. Potential Sabotage of Host Cell Physiology by Apicomplexan Parasites for Their Survival Benefits. Front Immunol 2017; 8:1261. [PMID: 29081773 PMCID: PMC5645534 DOI: 10.3389/fimmu.2017.01261] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/21/2017] [Indexed: 12/26/2022] Open
Abstract
Plasmodium, Toxoplasma, Cryptosporidium, Babesia, and Theileria are the major apicomplexan parasites affecting humans or animals worldwide. These pathogens represent an excellent example of host manipulators who can overturn host signaling pathways for their survival. They infect different types of host cells and take charge of the host machinery to gain nutrients and prevent itself from host attack. The mechanisms by which these pathogens modulate the host signaling pathways are well studied for Plasmodium, Toxoplasma, Cryptosporidium, and Theileria, except for limited studies on Babesia. Theileria is a unique pathogen taking into account the way it modulates host cell transformation, resulting in its clonal expansion. These parasites majorly modulate similar host signaling pathways, however, the disease outcome and effect is different among them. In this review, we discuss the approaches of these apicomplexan to manipulate the host–parasite clearance pathways during infection, invasion, survival, and egress.
Collapse
Affiliation(s)
| | - Sonti Roy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Hiral Uday Mistry
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Shweta Murthy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Neena George
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | | | - Paresh Sharma
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| |
Collapse
|
45
|
Lai MY, Lau YL. Screening and identification of host proteins interacting with Toxoplasma gondii SAG2 by yeast two-hybrid assay. Parasit Vectors 2017; 10:456. [PMID: 28969712 PMCID: PMC5625703 DOI: 10.1186/s13071-017-2387-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The identification of receptors or binding partners of Toxoplasma gondii from humans is an essential activity. Many proteins involved in T. gondii invasion have been characterized, and their contribution for parasite entry has been proposed. However, their molecular interactions remain unclear. RESULTS Yeast two-hybrid (Y2H) experiment was used to identify the binding partners of surface antigens of T. gondii by using SAG2 as bait. Colony PCR was performed and positive clones were sent for sequencing to confirm their identity. The yeast plasmids for true positive clones were rescued by transformation into E. coli TOP 10F' cells. The interplay between bait and prey was confirmed by β-galactosidase assay and co-immunoprecipitation experiment. We detected 20 clones interacting with SAG2 based on a series of the selection procedures. Following the autoactivation and toxicity tests, SAG2 was proven to be a suitable candidate as a bait. Thirteen clones were further examined by small scale Y2H experiment. The results indicated that a strong interaction existed between Homo sapiens zinc finger protein and SAG2, which could activate the expressions of the reporter genes in diploid yeast. Co-immunoprecipitation experiment result indicated the binding between this prey and SAG2 protein was significant (Mann-Whitney U-test: Z = -1.964, P = 0.05). CONCLUSIONS Homo sapiens zinc finger protein was found to interact with SAG2. To improve the understanding of this prey protein's function, advanced investigations need to be carried out.
Collapse
Affiliation(s)
- Meng-Yee Lai
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yee-Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
46
|
Translational Control in the Latency of Apicomplexan Parasites. Trends Parasitol 2017; 33:947-960. [PMID: 28942109 DOI: 10.1016/j.pt.2017.08.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/08/2017] [Accepted: 08/14/2017] [Indexed: 01/07/2023]
Abstract
Apicomplexan parasites Toxoplasma gondii and Plasmodium spp. use latent stages to persist in the host, facilitate transmission, and thwart treatment of infected patients. Therefore, it is important to understand the processes driving parasite differentiation to and from quiescent stages. Here, we discuss how a family of protein kinases that phosphorylate the eukaryotic initiation factor-2 (eIF2) function in translational control and drive differentiation. This translational control culminates in reprogramming of the transcriptome to facilitate parasite transition towards latency. We also discuss how eIF2 phosphorylation contributes to the maintenance of latency and provides a crucial role in the timing of reactivation of latent parasites towards proliferative stages.
Collapse
|
47
|
He C, Chen AY, Wei HX, Feng XS, Peng HJ. Phosphoproteome of Toxoplasma gondii Infected Host Cells Reveals Specific Cellular Processes Predominating in Different Phases of Infection. Am J Trop Med Hyg 2017; 97:236-244. [PMID: 28719319 DOI: 10.4269/ajtmh.16-0901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The invasion of Toxoplasma gondii tachyzoites into the host cell results in extensive host cell signaling activation/deactivation that is usually regulated by the phosphorylation/dephosphorylation. To elucidate how T. gondii regulates host cell signal transduction, the comparative phosphoproteome of stable isotope labeling with amino acids in cell culture-labeled human foreskin fibroblast cells was analyzed. The cells were grouped (Light [L], Medium [M], and Heavy [H] groups) based on the labeling isotope weight and were infected with T. gondii for different lengths of time (L: 0 hour; M: 2 hours; and H: 6 hours). A total of 892 phosphoproteins were identified with 1,872 phosphopeptides and 1,619 phosphorylation sites. The M versus L comparison revealed 694 significantly regulated phosphopeptides (436 upregulated and 258 downregulated). The H versus L comparison revealed 592 significantly regulated phosphopeptides (146 upregulated and 446 downregulated). The H versus M comparison revealed 794 significantly regulated phosphopeptides (149 upregulated and 645 downregulated). At 2 and 6 hours post-T. gondii infection, the most predominant host cell reactions were cell cycle regulation and cytoskeletal reorganization, which might be required for the efficient invasion and multiplication of T. gondii. Similar biological process profiles but different molecular function categories of host cells infected with T. gondii for 2 and 6 hours, which suggested that the host cell processes were not affected significantly by T. gondii infection but emphasized some differences in specific cellular processes at this two time points. Western blotting verification of some significantly regulated phosphoprotein phosphorylation sites was consistent with the mass spectra data. This study provided new insights into and further understanding of pathogen-host interactions from the host cell perspective.
Collapse
Affiliation(s)
- Cheng He
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, and Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Ai-Yuan Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, and Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Hai-Xia Wei
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, and Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Xiao-Shuang Feng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, and Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Hong-Juan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, and Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| |
Collapse
|
48
|
Xue J, Jiang W, Chen Y, Gong F, Wang M, Zeng P, Xia C, Wang Q, Huang K. Thioredoxin reductase from Toxoplasma gondii: an essential virulence effector with antioxidant function. FASEB J 2017; 31:4447-4457. [PMID: 28687608 DOI: 10.1096/fj.201700008r] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/12/2017] [Indexed: 12/20/2022]
Abstract
Thioredoxin reductase (TR) can help pathogens resist oxidative-burst injury from host immune cells by maintaining a thioredoxin-reduction state during NADPH consumption. TR is a necessary virulence factor that enables the persistent infection of some parasites. We performed bioinformatics analyses and biochemical assays to characterize the activity, subcellular localization, and genetic ablation of Toxoplasma gondii TR (TgTR), to shed light on its biologic function. We expressed the TgTR protein with an Escherichia coli expression system and analyzed its enzyme activity, reporting a Km for the recombinant TgTR of 11.47-15.57 μM, using NADPH as a substrate, and 130.48-151.09 μM with dithio-bis-nitrobenzoic acid as a substrate. The TgTR sequence shared homology with that of TR, but lacked a selenocysteine residue in the C-terminal region and was thought to contain 2 flavin adenine dinucleotide (FAD) domains and 1 NADPH domain. In addition, immunoelectron microscopy results showed that TgTR was widely dispersed in the cytoplasm, and we observed that parasite antioxidant capacity, invasion efficiency, and proliferation were decreased in TR-knockout (TR-KO) strains in vitro, although this strain still stimulated the release of reactive oxygen species release in mouse macrophages while being more sensitive to H2O2 toxicity in vitro Furthermore, our in vivo results revealed that the survival time of mice infected with the TR-KO strain was significantly prolonged relative to that of mice infected with the wild-type strain. These results suggest that TgTR plays an important role in resistance to oxidative damage and can be considered a virulence factor associated with T. gondii infection.-Xue, J., Jiang, W., Chen, Y., Gong, F., Wang, M., Zeng, P., Xia, C., Wang, Q., Huang, K. Thioredoxin reductase from Toxoplasma gondii: an essential virulence effector with antioxidant function.
Collapse
Affiliation(s)
- Junxin Xue
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China.,College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Shanghai Entry-Exit Inspection and Quarantine Bureau, Shanghai, China
| | - Wei Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Yongjun Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Fengju Gong
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Minyan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Peng Zeng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Can Xia
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Quan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China;
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China;
| |
Collapse
|
49
|
Cabral CM, McGovern KE, MacDonald WR, Franco J, Koshy AA. Dissecting Amyloid Beta Deposition Using Distinct Strains of the Neurotropic Parasite Toxoplasma gondii as a Novel Tool. ASN Neuro 2017; 9:1759091417724915. [PMID: 28817954 PMCID: PMC5565021 DOI: 10.1177/1759091417724915] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 01/13/2023] Open
Abstract
Genetic and pathologic data suggest that amyloid beta (Aβ), produced by processing of the amyloid precursor protein, is a major initiator of Alzheimer's disease (AD). To gain new insights into Aβ modulation, we sought to harness the power of the coevolution between the neurotropic parasite Toxoplasma gondii and the mammalian brain. Two prior studies attributed Toxoplasma-associated protection against Aβ to increases in anti-inflammatory cytokines (TGF-β and IL-10) and infiltrating phagocytic monocytes. These studies only used one Toxoplasma strain making it difficult to determine if the noted changes were associated with Aβ protection or simply infection. To address this limitation, we infected a third human amyloid precursor protein AD mouse model (J20) with each of the genetically distinct, canonical strains of Toxoplasma (Type I, Type II, or Type III). We then evaluated the central nervous system (CNS) for Aβ deposition, immune cell responses, global cytokine environment, and parasite burden. We found that only Type II infection was protective against Aβ deposition despite both Type II and Type III strains establishing a chronic CNS infection and inflammatory response. Compared with uninfected and Type I-infected mice, both Type II- and Type III-infected mice showed increased numbers of CNS T cells and microglia and elevated pro-inflammatory cytokines, but neither group showed a >2-fold elevation of TGF-β or IL-10. These data suggest that we can now use our identification of protective (Type II) and nonprotective (Type III) Toxoplasma strains to determine what parasite and host factors are linked to decreased Aβ burden rather than simply with infection.
Collapse
Affiliation(s)
| | | | - Wes R. MacDonald
- Undergraduate Biology Research Program, University of Arizona, Tucson, AZ, USA
| | - Jenna Franco
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Anita A. Koshy
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
- Department of Neurology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
50
|
Evolution of the Apicomplexan Sugar Transporter Gene Family Repertoire. Int J Genomics 2017; 2017:1707231. [PMID: 28555190 PMCID: PMC5438862 DOI: 10.1155/2017/1707231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/30/2017] [Indexed: 11/17/2022] Open
Abstract
Apicomplexan protist parasites utilize host sugars transported into the parasite by sugar transporter proteins for use as an energy source. We performed a phylum-wide phylogenetic analysis of the apicomplexan sugar transporter repertoire. Phylogenetic analyses revealed six major subfamilies of apicomplexan sugar transporters. Transporters in one subfamily have undergone expansions in Piroplasma species and Gregarina niphandrodes, while other subfamilies are highly divergent and contain genes found in only one or two species. Analyses of the divergent apicomplexan subfamilies revealed their presence in ciliates, indicating their alveolate ancestry and subsequent loss in chromerids and many apicomplexans.
Collapse
|